1
|
Hooijmaijers L, Vidal-Manrique M, Spils B, van Ens D, Rodriguez VC, Hobo W, de Goede AL, van Dorp S, Jansen JH, van der Waart AB, de Jonge PKJD, Dolstra H. Good manufacturing practice production of an off-the-shelf CD34 + progenitor-derived NK cell product with preserved anti-tumor functionality post-infusion in NOD/SCID/IL2Rg null mice. Cell Mol Life Sci 2025; 82:210. [PMID: 40415098 DOI: 10.1007/s00018-025-05727-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 04/18/2025] [Accepted: 04/24/2025] [Indexed: 05/27/2025]
Abstract
The adoptive transfer of natural killer (NK) cells represents a promising cancer therapy due to their intrinsic ability to distinguish between malignant and healthy cells in an allogeneic context, enabling off-the-shelf manufacturing possibilities. On demand availability of cryopreserved advanced therapy medicinal products (ATMPs) could promote enrolment in clinical trials and eventually commercialization with repeated dosing possibilities. However, NK cells are considered highly sensitive to cryopreservation-induced defects, including impaired viability, anti-tumor cytotoxicity, and in vivo expansion capacity. Here, we present the GMP-compliant manufacturing of an off-the-shelf NK cell product (RNK001), derived ex vivo from CD34+ hematopoietic stem and progenitor cells (HSPCs). To facilitate scalability and reduce hands-on time, the production process was adapted to a G-rex bioreactor, yielding high numbers of a pure CD56+CD3- NK cell product. Cryopreservation of HSPC-NK cells using an optimized freeze-thaw protocol resulted in a consistently high post-thawing viability of mature and differentiated cells. Surviving HSPC-NK cells post-thawing exhibited enhanced proliferative capacity compared to fresh cells in vitro and their persistence in vivo was similar to that of fresh cells when administrated intravenously or intraperitoneally in NOD/SCID/IL2Rgnull mice. Moreover, cryopreserved HSPC-NK cells had robust anti-tumor functionality, efficiently killing tumor spheroids embedded in a 3D collagen matrix and maintaining degranulation and interferon-γ production capacity comparable to fresh cells following in vivo infusion. Together, these findings show the potential of cryopreserved HSPC-NK cells with potent effector functions, allowing the manufacturing of an off-the-shelf therapeutical NK cell product for hematological and solid malignancies.
Collapse
Affiliation(s)
- Laura Hooijmaijers
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marcos Vidal-Manrique
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bart Spils
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Diede van Ens
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Verónica Castaño Rodriguez
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Willemijn Hobo
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anna L de Goede
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Suzanne van Dorp
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joop H Jansen
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anniek B van der Waart
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Paul K J D de Jonge
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Harry Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
2
|
Ji S, Jin C, Cui X. Enhancing the physiological characteristics of chimeric antigen receptor natural killer cells by synthetic biology. Front Immunol 2025; 16:1592121. [PMID: 40313937 PMCID: PMC12043574 DOI: 10.3389/fimmu.2025.1592121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Accepted: 04/02/2025] [Indexed: 05/03/2025] Open
Abstract
Chimeric antigen receptor natural Killer (CAR-NK) cells therapy represents a next-generation immunotherapeutic approach following CAR-T cells therapy, offering inherent "off-the-shelf" compatibility and mitigated off-tumor toxicity. Despite these advantages, clinical translation remains constrained by poor in vivo persistence and functional exhaustion in immunosuppressive tumor microenvironments (TME). This review examines recent advancements in synthetic biology aimed at enhancing the physiological characteristics of CAR-NK cells. By delineating the synergy between NK cells and synthetic biology toolkits, this work provides a roadmap for developing next-generation CAR-NK therapies capable of addressing solid tumor challenges while maintaining favorable safety profiles.
Collapse
Affiliation(s)
- Shuochao Ji
- Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Cheng Jin
- Department of Hematology and Oncology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Xinjiang Cui
- Affiliated Hospital of Shandong Second Medical University, Weifang, China
| |
Collapse
|
3
|
Wang Y, Li Y, Chen J, Guo C, Yu X, Zhang Z, Fu Y, Han X, Hu Q, Ding H, Shang H, Jiang Y. Inhibition of TIGIT on NK cells improves their cytotoxicity and HIV reservoir eradication potential. mBio 2025; 16:e0322624. [PMID: 39918313 PMCID: PMC11898710 DOI: 10.1128/mbio.03226-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/10/2025] [Indexed: 03/14/2025] Open
Abstract
The latent human immunodeficiency virus (HIV) reservoir presents the biggest obstacle to curing HIV chronic infection. Consequently, finding novel strategies to control the HIV reservoir is critical. Natural killer (NK) cells are essential for antiviral immunity. However, the influence of NK cell subsets and their associated inhibitory or activating receptors on their cytotoxicity toward the HIV reservoir has not been fully studied. We investigated the relationship between the percentage of NK cells or NK cell subsets and the HIV reservoir. Our results indicated that the percentage of CD56-CD16+ NK cells was positively associated with HIV reservoir size (i.e., HIV DNA, HIV msRNA, or HIV usRNA). Additionally, we observed that the percentage of IFN-γ+ NK cells was inversely related to the HIV reservoir. Furthermore, the expression of TIGIT on NK cells, particularly CD56-CD16+ and CD56dim NK cell subsets, positively correlated with the HIV reservoir. Notably, individuals with higher percentage of TIGIT+ NK and lower percentage of CD226+ NK cells exhibited larger HIV reservoir. Mechanistically, we discovered that TIGIT could inhibit the PI3K-Akt-mTOR-mTORC1 (s6k) signaling pathway to decrease the production of IFN-γ in NK cells. Importantly, inhibiting TIGIT in NK cells enhanced their ability to eliminate reactivated latently infected CD4+ T cells. Our experiments underscored the crucial role of NK cells in controlling the HIV reservoir and suggested that TIGIT serves as a promising target for enhancing the NK cell-mediated clearance of the HIV reservoir. IMPORTANCE As a major barrier to human immunodeficiency virus (HIV) cure, HIV reservoir persist in viremia-suppressed infected individuals. NK cells are important antiviral cells, and their impact on reservoir has rarely been reported. We analyzed the relationship between the size of reservoir and NK cell subsets, inhibitory receptor TIGIT expression. Our analysis found that the percentage of CD56-CD16+ NK cells was positively associated with HIV reservoir size. Furthermore, TIGIT expression on NK cells and CD56-CD16+ NK cells or CD56dim NK cells has a positive correlation with the HIV reservoir. TIGIT can inhibit the PI3K-Akt-mTOR-mTORC1 (s6k) signaling pathway to decrease the production of IFN-γ on NK cells. Blocking TIGIT in NK cells can enhance their ability to eliminate reactivated latently infected CD4+ T cells. Our study indicated that NK cells are critical to the control of the reservoir size, and TIGIT may be a target for enhancing the NK cell-mediated elimination of the reservoir.
Collapse
Affiliation(s)
- Yue Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, China
- Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China
- Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, China
| | - Yidi Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, China
- Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China
- Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, China
| | - Jiaqi Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, China
- Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China
- Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, China
| | - Chenxi Guo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, China
- Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China
- Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, China
| | - Xiaowen Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, China
- Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China
- Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, China
| | - Zining Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, China
- Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China
- Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, China
| | - Yajing Fu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, China
- Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China
- Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, China
| | - Xiaoxu Han
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, China
- Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China
- Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, China
| | - Qinghai Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, China
- Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China
- Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, China
| | - Haibo Ding
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, China
- Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China
- Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, China
| | - Hong Shang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, China
- Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China
- Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Yongjun Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, China
- Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, China
- Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, China
| |
Collapse
|
4
|
Song Y, Lei L, Cai X, Wei H, Yu CY. Immunomodulatory Peptides for Tumor Treatment. Adv Healthc Mater 2025; 14:e2400512. [PMID: 38657003 DOI: 10.1002/adhm.202400512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/07/2024] [Indexed: 04/26/2024]
Abstract
Peptides exhibit various biological activities, including biorecognition, cell targeting, and tumor penetration, and can stimulate immune cells to elicit immune responses for tumor immunotherapy. Peptide self-assemblies and peptide-functionalized nanocarriers can reduce the effect of various biological barriers and the degradation by peptidases, enhancing the efficiency of peptide delivery and improving antitumor immune responses. To date, the design and development of peptides with various functionalities have been extensively reviewed for enhanced chemotherapy; however, peptide-mediated tumor immunotherapy using peptides acting on different immune cells, to the knowledge, has not yet been summarized. Thus, this work provides a review of this emerging subject of research, focusing on immunomodulatory anticancer peptides. This review introduces the role of peptides in the immunomodulation of innate and adaptive immune cells, followed by a link between peptides in the innate and adaptive immune systems. The peptides are discussed in detail, following a classification according to their effects on different innate and adaptive immune cells, as well as immune checkpoints. Subsequently, two delivery strategies for peptides as drugs are presented: peptide self-assemblies and peptide-functionalized nanocarriers. The concluding remarks regarding the challenges and potential solutions of peptides for tumor immunotherapy are presented.
Collapse
Affiliation(s)
- Yang Song
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Longtianyang Lei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xingyu Cai
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Affiliated Hospital of Hunan Academy of Chinese Medicine, Hunan Academy of Chinese Medicine, Changsha, 410013, China
| |
Collapse
|
5
|
Nguyen TTT, Jang B, Kim SR, Kang SK, Kim KY, Kim YH, Koh YH. Enhanced Immune Functions of In Vitro Human Natural Killer Cells and Splenocytes in Immunosuppressed Mice Supplemented with Mature Silkworm Products. Nutrients 2025; 17:417. [PMID: 39940275 PMCID: PMC11820884 DOI: 10.3390/nu17030417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/12/2025] [Accepted: 01/22/2025] [Indexed: 02/14/2025] Open
Abstract
OBJECTIVES The immune-enhancing properties of steamed mature silkworm, known as HongJam (HJ), were investigated using human interleukin-2-independent Natural Killer 92 (NK92-MI) cells and a cyclophosphamide intraperitoneal injection-induced immunosuppressed mice model (CPA-IP). White Jade variety mature silkworm HJ (WJ-HJ) was used to prepare WJ-HJ supercritical fluid extracts (WJ-SCE) and WJ-HJ-supplemented feeds. RESULTS Treatment with WJ-SCE significantly enhanced proliferation, migration, and cytotoxicity of NK92-MI cells against various cancer cells while improving mitochondrial function and ATP production (p < 0.05). In CPA-IP mice, consumption of WJ-HJ-supplemented feeds restored immune function by improving body weight, immune organ indices, immunoglobulin levels, and blood cytokines. Splenocyte proliferation and cytotoxicity were significantly elevated in both saline intraperitoneal injection (Sal-IP) and CPA-IP groups with WJ-HJ supplementation, independent of mitogen activation (p < 0.05). CONCLUSIONS These results suggest that WJ-HJ enhances immune modulation and immune surveillance functions of NK cells by improving mitochondrial and cytotoxic functions. WJ-HJ holds promise as a functional food for immune enhancement, pending clinical validation.
Collapse
Affiliation(s)
- Thanh Thi Tam Nguyen
- Department of Biomedical Gerontology, Hallym University Graduate School, Chuncheon 24252, Republic of Korea;
| | - Byungki Jang
- Ilsong Institute of Life Science, Hallym University, Seoul 07441, Republic of Korea; (B.J.); (Y.H.K.)
| | - Seong-Ruyl Kim
- Division of Industrial Insects and Sericulture, National Institute of Agricultural Sciences, Wanju 55365, Republic of Korea; (S.-R.K.); (S.-K.K.); (K.-Y.K.)
| | - Sang-Kuk Kang
- Division of Industrial Insects and Sericulture, National Institute of Agricultural Sciences, Wanju 55365, Republic of Korea; (S.-R.K.); (S.-K.K.); (K.-Y.K.)
| | - Kee-Young Kim
- Division of Industrial Insects and Sericulture, National Institute of Agricultural Sciences, Wanju 55365, Republic of Korea; (S.-R.K.); (S.-K.K.); (K.-Y.K.)
| | - Yoo Hee Kim
- Ilsong Institute of Life Science, Hallym University, Seoul 07441, Republic of Korea; (B.J.); (Y.H.K.)
| | - Young Ho Koh
- Department of Biomedical Gerontology, Hallym University Graduate School, Chuncheon 24252, Republic of Korea;
- Ilsong Institute of Life Science, Hallym University, Seoul 07441, Republic of Korea; (B.J.); (Y.H.K.)
| |
Collapse
|
6
|
Rados M, Landegger A, Schmutzler L, Rabidou K, Taschner-Mandl S, Fetahu IS. Natural killer cells in neuroblastoma: immunological insights and therapeutic perspectives. Cancer Metastasis Rev 2024; 43:1401-1417. [PMID: 39294470 PMCID: PMC11554946 DOI: 10.1007/s10555-024-10212-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 09/10/2024] [Indexed: 09/20/2024]
Abstract
Natural killer (NK) cells have multifaceted roles within the complex tumor milieu. They are pivotal components of innate immunity and shape the dynamic landscape of tumor-immune cell interactions, and thus can be leveraged for use in therapeutic interventions. NK-based immunotherapies have had remarkable success in hematological malignancies, but these therapies are met with many challenges in solid tumors, including neuroblastoma (NB), a childhood tumor arising from the sympathetic nervous system. With a focus on NB, this review outlines the mechanisms employed by NK cells to recognize and eliminate malignant cells, delving into the dynamic relationship between ligand-receptor interactions, cytokines, and other molecules that facilitate the cross talk between NK and NB cells. We discuss the immunomodulatory functions of NK cells and the mechanisms that contribute to loss of this immunosurveillance in NB, with a focus on how this dynamic has been utilized in recent immunotherapy advancements for NB.
Collapse
Affiliation(s)
- Magdalena Rados
- St. Anna Children's Cancer Research Institute, Vienna, Austria
| | | | - Lukas Schmutzler
- Department of Otorhinolaryngology - Head and Neck Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Kimberlie Rabidou
- Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, USA
| | | | - Irfete S Fetahu
- Department of Neurology, Division of Neuropathology and Neurochemistry, Medical University of Vienna, Vienna, Austria.
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
7
|
Katz AR, Huntwork MP, Kolls JK, Hewes JL, Ellsworth CR, Clark RDE, Carlson JC. Impact of psychological stressors on natural killer cell function: A comprehensive analysis based on stressor type, duration, intensity, and species. Physiol Behav 2024; 288:114734. [PMID: 39547436 DOI: 10.1016/j.physbeh.2024.114734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 09/11/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
Patients with natural killer (NK) cell deficiency or dysfunction are more susceptible to infections by Herpesviridae viruses, herpesvirus-related cancers, and macrophage activation syndromes. This review summarizes research on NK cell dysfunction following psychological stress, focusing on stressor type, duration, age of exposure, and species studied. Psychological stressors negatively affect NK cell activity (NKCA) across species. Prolonged stress leads to more significant decreases in NK cell number and function, with rehabilitation efforts proving ineffective in reversing these effects. Early life and prolonged stress exposure particularly increases the risk of infections and cancer due to impaired NKCA. The review also highlights that stress impacts males and females differently, with females exhibiting a more immunosuppressed NK cell phenotype. Notably, mice respond differently compared to humans and other animals, making them unsuitable for NK cell stress-related studies. Most studies measured NKCA using cytolytic assays against K-562 or YAC-1 cells. Although the exact mechanisms of NK cell dysfunction under stress remain unclear, potential causes include reduced release of secretory lysosomes with perforin or granzyme, impaired NK cell synapse formation, decreased expression of synapse-related molecules like CD2 or LFA-1 (CD11a), altered activating receptor expression, and dysregulated signaling pathways, such as decreased Erk1/2 phosphorylation and NFkB signaling. These mechanisms are not mutually exclusive, and future research is needed to clarify these pathways and develop therapeutic interventions for stress-induced immune dysregulation.
Collapse
Affiliation(s)
- Alexis R Katz
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA; Departments of Medicine and Pediatrics, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| | - Margaret P Huntwork
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA; Department of Allergy and Clinical Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jay K Kolls
- Departments of Medicine and Pediatrics, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jenny L Hewes
- Departments of Medicine and Pediatrics, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Calder R Ellsworth
- Departments of Medicine and Pediatrics, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Robert D E Clark
- Departments of Medicine and Pediatrics, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - John C Carlson
- Department of Allergy and Immunology, Ochsner Health System, New Orleans, LA 70121, USA
| |
Collapse
|
8
|
Almutairi A, Alshehri NA, Al Subayyil A, Bahattab E, Alshabibi M, Abomaray F, Basmaeil YS, Khatlani T. Human decidua basalis mesenchymal stem/stromal cells enhance anticancer properties of human natural killer cells, in vitro. Front Cell Dev Biol 2024; 12:1435484. [PMID: 39539962 PMCID: PMC11557523 DOI: 10.3389/fcell.2024.1435484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Mesenchymal stem cells/stromal cells from the Decidua Basalis of the human placenta (DBMSCs) express wide range of effector molecules that modulate the functions of their target cells. These properties make them potential candidate for use in cellular therapy. In this study, we have investigated the consequences of interaction between DBMSCs and natural killer (NK) cells for both cell types. Methods DBMSCs were cultured with IL-2-activated and resting non-activated NK cells isolated from healthy human peripheral blood and various functional assays were performed including, NK cell proliferation and cytolytic activities. Flow cytometry and microscopic studies were performed to examine the expression of NK cell receptors that mediate these cytolytic activities against DBMSCs. Moreover, the mechanism underlying these effects was also investigated. Results Our findings revealed that, co-culture of DBMSCs and NK cells resulted in inhibition of proliferation of resting NK cells, while proliferation of IL-2 activated NK cells was increased. Contrarily, treatment of DBMSC's with comparatively high numbers of IL-2 activated NK cells, resulted in their lysis, whereas treatment with low numbers resulted in reduction in their proliferation. Cytolytic activity of NK cells against DBMSCs was mediated by several activating NK cell receptors. In spite of the expression of HLA class I molecules by DBMSCs, they were still lysed by NK cells, excluding their involvement in cytolytic activity. In addition, preconditioning NK cells by DBMSCs, enhanced their ability to suppress tumor cell proliferation and in severe cases resulted in their partial lysis. Lysis and decrease of tumor cell proliferation is associated with increased expression of important molecules involved in anticancer activities. Discussion We conclude that DBMSCs exhibit dualfunctions on NK cells that enhance their anticancer therapeutic potential.
Collapse
Affiliation(s)
- Abdulaziz Almutairi
- College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdul Aziz University for Medical Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh, Saudi Arabia
| | - Najlaa A. Alshehri
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdul Aziz University for Medical Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh, Saudi Arabia
- School of Education, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Abdullah Al Subayyil
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdul Aziz University for Medical Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh, Saudi Arabia
| | - Eman Bahattab
- National Center for Stem Cell Technology, Life Sciences and Environment Research Institute, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Manal Alshabibi
- National Center for Stem Cell Technology, Life Sciences and Environment Research Institute, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Fawaz Abomaray
- Department of Clinical Science, Intervention and Technology, Division of Obstetrics and Gynecology, Karolinska Institutet, Stockholm, Sweden
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Yasser S. Basmaeil
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdul Aziz University for Medical Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh, Saudi Arabia
| | - Tanvir Khatlani
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdul Aziz University for Medical Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh, Saudi Arabia
| |
Collapse
|
9
|
Sun Y, Pan Z, Wang Z, Wang H, Wei L, Cui F, Zou Q, Zhang Z. Single-cell transcriptome analysis reveals immune microenvironment changes and insights into the transition from DCIS to IDC with associated prognostic genes. J Transl Med 2024; 22:894. [PMID: 39363164 PMCID: PMC11448450 DOI: 10.1186/s12967-024-05706-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Ductal carcinoma in situ (DCIS) of the breast is an early stage of breast cancer, and preventing its progression to invasive ductal carcinoma (IDC) is crucial for the early detection and treatment of breast cancer. Although single-cell transcriptome analysis technology has been widely used in breast cancer research, the biological mechanisms underlying the transition from DCIS to IDC remain poorly understood. RESULTS We identified eight cell types through cell annotation, finding significant differences in T cell proportions between DCIS and IDC. Using this as a basis, we performed pseudotime analysis on T cell subpopulations, revealing that differentially expressed genes primarily regulate immune cell migration and modulation. By intersecting WGCNA results of T cells highly correlated with the subtypes and the differentially expressed genes, we identified six key genes: FGFBP2, GNLY, KLRD1, TYROBP, PRF1, and NKG7. Excluding PRF1, the other five genes were significantly associated with overall survival in breast cancer, highlighting their potential as prognostic biomarkers. CONCLUSIONS We identified immune cells that may play a role in the progression from DCIS to IDC and uncovered five key genes that can serve as prognostic markers for breast cancer. These findings provide insights into the mechanisms underlying the transition from DCIS to IDC, offering valuable perspectives for future research. Additionally, our results contribute to a better understanding of the biological processes involved in breast cancer progression.
Collapse
MESH Headings
- Humans
- Single-Cell Analysis
- Female
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
- Gene Expression Profiling
- Prognosis
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Carcinoma, Intraductal, Noninfiltrating/immunology
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Breast Neoplasms/genetics
- Breast Neoplasms/immunology
- Breast Neoplasms/pathology
- Gene Expression Regulation, Neoplastic
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/immunology
- Disease Progression
- Transcriptome/genetics
- Single-Cell Gene Expression Analysis
Collapse
Affiliation(s)
- Yidi Sun
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China
| | - Zhuoyu Pan
- International Business School, Hainan University, Haikou, 570228, China
| | - Ziyi Wang
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China
| | - Haofei Wang
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China
| | - Leyi Wei
- Centre for Artificial Intelligence driven Drug Discovery, Faculty of Applied Science, Macao Polytechnic University, Macao SAR, China
- School of Informatics, Xiamen University, Xiamen, China
| | - Feifei Cui
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China.
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610054, China.
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, 324000, China.
| | - Zilong Zhang
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China.
| |
Collapse
|
10
|
Mahdifar M, Boostani R, Taylor GP, Rezaee SA, Rafatpanah H. Comprehensive Insight into the Functional Roles of NK and NKT Cells in HTLV-1-Associated Diseases and Asymptomatic Carriers. Mol Neurobiol 2024; 61:7877-7889. [PMID: 38436833 DOI: 10.1007/s12035-024-03999-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 01/29/2024] [Indexed: 03/05/2024]
Abstract
Human T cell leukemia virus type 1 (HTLV-1) is the first human oncogenic retrovirus to be discovered and causes two major diseases: a progressive neuro-inflammatory disease, termed HTLV-1 associated myelopathy/tropical spastic paraparesis (HAM/TSP), and an aggressive malignancy of T lymphocytes known as adult T cell leukemia (ATL). Innate and acquired immune responses play pivotal roles in controlling the status of HTLV-1-infected cells and such, the outcome of HTLV-1 infection. Natural killer cells (NKCs) are the effector cells of the innate immune system and are involved in controlling viral infections and several types of cancers. The ability of NKCs to trigger cytotoxicity to provide surveillance against viruses and cancer depends on the balance between the inhibitory and activating signals. In this review, we will discuss NKC function and the alterations in the frequency of these cells in HTLV-1 infection.
Collapse
Affiliation(s)
- Maryam Mahdifar
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Boostani
- Department of Neurology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Graham P Taylor
- Section of Infectious Diseases, Department of Medicine, Imperial College London, London, UK
| | - Seyed Abdolrahim Rezaee
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Houshang Rafatpanah
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
11
|
Tao S, You X, Norman PJ, Kichula KM, Dong L, Chen N, He J, Zhang W, Zhu F. Analysis of KIR and HLA Polymorphism in Chinese Individuals With COVID-19. HLA 2024; 104:e15715. [PMID: 39364548 PMCID: PMC11458138 DOI: 10.1111/tan.15715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/03/2024] [Accepted: 09/17/2024] [Indexed: 10/05/2024]
Abstract
Killer-cell immunoglobulin-like receptor (KIR) interactions with HLA class I have crucial roles in modulating NK cell function in response to viral infections. To explore the correlation between KIR/HLA and susceptibility to SARS-CoV-2 infection, we analysed polymorphism of KIR genes, haplotypes, HLA allotypes, and the interplay between KIR and HLA in individuals diagnosed with COVID-19. Compared to a population control group, we observed a significantly increased frequency of KIR3DL3*00802 in the COVID-19 group. When encoded by the HLA-B gene, the frequency of HLA-Bw4, a ligand for KIR3DL1, was at lower frequency in the COVID-19 group. Additionally, significantly elevated frequencies of KIR-Bx3, KIR3DL3*00301, 3DL3*048, and C1+HLA-C were identified in the COVID-19 group before multiple test correction, suggesting associations with susceptibility to SARS-CoV-2 infection. Our findings indicate that the KIR3DL3*00802 allele may be a high-risk factor for SARS-CoV-2 infection, while Bw4 encoded by HLA-B gene may confer protective effects against the infection.
Collapse
Affiliation(s)
- Sudan Tao
- Blood Center of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Xuan You
- Blood Center of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Paul J. Norman
- Department of Biomedical Informatics and Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Katherine M. Kichula
- Department of Biomedical Informatics and Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Lina Dong
- Blood Center of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Nanying Chen
- Blood Center of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Ji He
- Blood Center of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Wei Zhang
- Blood Center of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Faming Zhu
- Blood Center of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
12
|
Martin KE, Hammer Q, Perica K, Sadelain M, Malmberg KJ. Engineering immune-evasive allogeneic cellular immunotherapies. Nat Rev Immunol 2024; 24:680-693. [PMID: 38658708 DOI: 10.1038/s41577-024-01022-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2024] [Indexed: 04/26/2024]
Abstract
Allogeneic cellular immunotherapies hold a great promise for cancer treatment owing to their potential cost-effectiveness, scalability and on-demand availability. However, immune rejection of adoptively transferred allogeneic T and natural killer (NK) cells is a substantial obstacle to achieving clinical responses that are comparable to responses obtained with current autologous chimeric antigen receptor T cell therapies. In this Perspective, we discuss strategies to confer cell-intrinsic, immune-evasive properties to allogeneic T cells and NK cells in order to prevent or delay their immune rejection, thereby widening the therapeutic window. We discuss how common viral and cancer immune escape mechanisms can serve as a blueprint for improving the persistence of off-the-shelf allogeneic cell therapies. The prospects of harnessing genome editing and synthetic biology to design cell-based precision immunotherapies extend beyond programming target specificities and require careful consideration of innate and adaptive responses in the recipient that may curtail the biodistribution, in vivo expansion and persistence of cellular therapeutics.
Collapse
Affiliation(s)
- Karen E Martin
- Precision Immunotherapy Alliance, The University of Oslo, Oslo, Norway
- Department of Cancer Immunology, Institute for Cancer Research Oslo, Oslo University Hospital, Oslo, Norway
| | - Quirin Hammer
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Karlo Perica
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Cell Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michel Sadelain
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karl-Johan Malmberg
- Precision Immunotherapy Alliance, The University of Oslo, Oslo, Norway.
- Department of Cancer Immunology, Institute for Cancer Research Oslo, Oslo University Hospital, Oslo, Norway.
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
13
|
Mohammad Taheri M, Javan F, Poudineh M, Athari SS. Beyond CAR-T: The rise of CAR-NK cell therapy in asthma immunotherapy. J Transl Med 2024; 22:736. [PMID: 39103889 PMCID: PMC11302387 DOI: 10.1186/s12967-024-05534-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/23/2024] [Indexed: 08/07/2024] Open
Abstract
Asthma poses a major public health burden. While existing asthma drugs manage symptoms for many, some patients remain resistant. The lack of a cure, especially for severe asthma, compels exploration of novel therapies. Cancer immunotherapy successes with CAR-T cells suggest its potential for asthma treatment. Researchers are exploring various approaches for allergic diseases including membrane-bound IgE, IL-5, PD-L2, and CTLA-4 for asthma, and Dectin-1 for fungal asthma. NK cells offer several advantages over T cells for CAR-based immunotherapy. They offer key benefits: (1) HLA compatibility, meaning they can be used in a wider range of patients without the need for matching tissue types. (2) Minimal side effects (CRS and GVHD) due to their limited persistence and cytokine profile. (3) Scalability for "off-the-shelf" production from various sources. Several strategies have been introduced that highlight the superiority and challenges of CAR-NK cell therapy for asthma treatment including IL-10, IFN-γ, ADCC, perforin-granzyme, FASL, KIR, NCRs (NKP46), DAP, DNAM-1, TGF-β, TNF-α, CCL, NKG2A, TF, and EGFR. Furthermore, we advocate for incorporating AI for CAR design optimization and CRISPR-Cas9 gene editing technology for precise gene manipulation to generate highly effective CAR constructs. This review will delve into the evolution and production of CAR designs, explore pre-clinical and clinical studies of CAR-based therapies in asthma, analyze strategies to optimize CAR-NK cell function, conduct a comparative analysis of CAR-T and CAR-NK cell therapy with their respective challenges, and finally present established novel CAR designs with promising potential for asthma treatment.
Collapse
Affiliation(s)
| | - Fatemeh Javan
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohadeseh Poudineh
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Seyed Shamseddin Athari
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
- Department of Immunology, Zanjan School of Medicine, Zanjan University of Medical Sciences, 12th Street, Shahrake Karmandan, Zanjan, 45139-561111, Iran.
| |
Collapse
|
14
|
Wang Q, Chen S, Guo Z, Xia S, Zhang M. NK-like CD8 T cell: one potential evolutionary continuum between adaptive memory and innate immunity. Clin Exp Immunol 2024; 217:136-150. [PMID: 38651831 PMCID: PMC11239564 DOI: 10.1093/cei/uxae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 03/06/2024] [Accepted: 04/22/2024] [Indexed: 04/25/2024] Open
Abstract
CD8 T cells are crucial adaptive immune cells with cytotoxicity to fight against pathogens or abnormal self-cells via major histocompatibility complex class I-dependent priming pathways. The composition of the memory CD8 T-cell pool is influenced by various factors. Physiological aging, chronic viral infection, and autoimmune diseases promote the accumulation of CD8 T cells with highly differentiated memory phenotypes. Accumulating studies have shown that some of these memory CD8 T cells also exhibit innate-like cytotoxicity and upregulate the expression of receptors associated with natural killer (NK) cells. Further analysis shows that these NK-like CD8 T cells have transcriptional profiles of both NK and CD8 T cells, suggesting the transformation of CD8 T cells into NK cells. However, the specific induction mechanism underlying NK-like transformation and the implications of this process for CD8 T cells are still unclear. This review aimed to deduce the possible differentiation model of NK-like CD8 T cells, summarize the functions of major NK-cell receptors expressed on these cells, and provide a new perspective for exploring the role of these CD8 T cells in health and disease.
Collapse
Affiliation(s)
- Qiulei Wang
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shaodan Chen
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zhenhong Guo
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Minghui Zhang
- School of Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
15
|
Sabag B, Puthenveetil A, Levy M, Joseph N, Doniger T, Yaron O, Karako-Lampert S, Lazar I, Awwad F, Ashkenazi S, Barda-Saad M. Dysfunctional natural killer cells can be reprogrammed to regain anti-tumor activity. EMBO J 2024; 43:2552-2581. [PMID: 38637625 PMCID: PMC11217363 DOI: 10.1038/s44318-024-00094-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 04/20/2024] Open
Abstract
Natural killer (NK) cells are critical to the innate immune system, as they recognize antigens without prior sensitization, and contribute to the control and clearance of viral infections and cancer. However, a significant proportion of NK cells in mice and humans do not express classical inhibitory receptors during their education process and are rendered naturally "anergic", i.e., exhibiting reduced effector functions. The molecular events leading to NK cell anergy as well as their relation to those underlying NK cell exhaustion that arises from overstimulation in chronic conditions, remain unknown. Here, we characterize the "anergic" phenotype and demonstrate functional, transcriptional, and phenotypic similarities to the "exhausted" state in tumor-infiltrating NK cells. Furthermore, we identify zinc finger transcription factor Egr2 and diacylglycerol kinase DGKα as common negative regulators controlling NK cell dysfunction. Finally, experiments in a 3D organotypic spheroid culture model and an in vivo tumor model suggest that a nanoparticle-based delivery platform can reprogram these dysfunctional natural killer cell populations in their native microenvironment. This approach may become clinically relevant for the development of novel anti-tumor immunotherapeutic strategies.
Collapse
Affiliation(s)
- Batel Sabag
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Abhishek Puthenveetil
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Moria Levy
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Noah Joseph
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Tirtza Doniger
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Orly Yaron
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Sarit Karako-Lampert
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Itay Lazar
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Fatima Awwad
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Shahar Ashkenazi
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Mira Barda-Saad
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel.
| |
Collapse
|
16
|
Shah Z, Tian L, Li Z, Jin L, Zhang J, Li Z, Barr T, Tang H, Feng M, Caligiuri MA, Yu J. Human anti-PSCA CAR macrophages possess potent antitumor activity against pancreatic cancer. Cell Stem Cell 2024; 31:803-817.e6. [PMID: 38663406 PMCID: PMC11162318 DOI: 10.1016/j.stem.2024.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 01/11/2024] [Accepted: 03/28/2024] [Indexed: 05/15/2024]
Abstract
Due to the limitations of autologous chimeric antigen receptor (CAR)-T cells, alternative sources of cellular immunotherapy, including CAR macrophages, are emerging for solid tumors. Human induced pluripotent stem cells (iPSCs) offer an unlimited source for immune cell generation. Here, we develop human iPSC-derived CAR macrophages targeting prostate stem cell antigen (PSCA) (CAR-iMacs), which express membrane-bound interleukin (IL)-15 and truncated epidermal growth factor receptor (EGFR) for immune cell activation and a suicide switch, respectively. These allogeneic CAR-iMacs exhibit strong antitumor activity against human pancreatic solid tumors in vitro and in vivo, leading to reduced tumor burden and improved survival in a pancreatic cancer mouse model. CAR-iMacs appear safe and do not exhibit signs of cytokine release syndrome or other in vivo toxicities. We optimized the cryopreservation of CAR-iMac progenitors that remain functional upon thawing, providing an off-the-shelf, allogeneic cell product that can be developed into CAR-iMacs. Overall, our preclinical data strongly support the potential clinical translation of this human iPSC-derived platform for solid tumors, including pancreatic cancer.
Collapse
Affiliation(s)
- Zahir Shah
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Lei Tian
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Zhixin Li
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Lewei Jin
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Jianying Zhang
- Department of Computational and Quantitative Medicine, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Zhenlong Li
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Tasha Barr
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Hejun Tang
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Mingye Feng
- Department of Immuno-Oncology, City of Hope, Los Angeles, CA 91010, USA
| | - Michael A Caligiuri
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA; City of Hope Comprehensive Cancer Center, Los Angeles, CA 91010, USA.
| | - Jianhua Yu
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Department of Immuno-Oncology, City of Hope, Los Angeles, CA 91010, USA; City of Hope Comprehensive Cancer Center, Los Angeles, CA 91010, USA.
| |
Collapse
|
17
|
Zhang H, Tang Q, Miao Y, Wang J, Yuan Z, Huang X, Zhu Y, Nong C, Li G, Cui R, Huang X, Zhang L, Yu Q, Jiang Z. Group 1 innate lymphoid cell activation via recognition of NKG2D and liver resident macrophage MULT-1: Collaborated roles in triptolide induced hepatic immunotoxicity in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 272:116072. [PMID: 38342011 DOI: 10.1016/j.ecoenv.2024.116072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/22/2024] [Accepted: 02/01/2024] [Indexed: 02/13/2024]
Abstract
Triptolide (TP) is the major bioactive component of traditional Chinese medicine Tripterygium wilfordii Hook. F., a traditional Chinese medicinal plant categorized within the Tripterygium genus of the Celastraceae family. It is recognized for its therapeutic potential in addressing a multitude of diseases. Nonetheless, TP is known to exhibit multi-organ toxicity, notably hepatotoxicity, which poses a significant concern for the well-being of patients undergoing treatment. The precise mechanisms responsible for TP-induced hepatotoxicity remain unresolved. In our previous investigation, it was determined that TP induces heightened hepatic responsiveness to exogenous lipopolysaccharide (LPS). Additionally, natural killer (NK) cells were identified as a crucial effector responsible for mediating hepatocellular damage in this context. However, associated activating receptors and the underlying mechanisms governing NK cell represented innate lymphoid cell (ILC) activation remained subjects of inquiry and were not yet investigated. Herein, activating receptor Killer cell lectin like receptor K1 (NKG2D) of group 1 ILCs was specifically upregulated in TP- and LPS-induced acute liver failure (ALF), and in vivo blockade of NKG2D significantly reduced group 1 ILC mediated cytotoxicity and mitigated TP- and LPS-induced ALF. NKG2D ligand UL16-binding protein-like transcript 1 (MULT-1) was found upregulated in liver resident macrophages (LRMs) after TP administration, and LRMs did exhibit NK cell activating effect. Furthermore, M1 polarization of LRMs cells was observed, along with an elevation in intracellular tumor necrosis factor (TNF)-α levels. In vivo neutralization of TNF-α significantly alleviated TP- and LPS-induced ALF. In conclusion, the collaborative role of group 1 ILCs and LRMs in mediating hepatotoxicity was confirmed in TP- and LPS-induced ALF. TP-induced MULT-1 expression in LRMs was the crucial mechanism in the activation of group 1 ILCs via MULT-1-NKG2D signal upon LPS stimulation, emphasizing the importance of infection control after TP administration.
Collapse
Affiliation(s)
- Haoran Zhang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Qianhui Tang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yingying Miao
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Jie Wang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Zihang Yuan
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Xinliang Huang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Ying Zhu
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Cheng Nong
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Guoqing Li
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ruyu Cui
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Xin Huang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Luyong Zhang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Qinwei Yu
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Zhenzhou Jiang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
18
|
Fan Z, Han D, Fan X, Zhao L. Ovarian cancer treatment and natural killer cell-based immunotherapy. Front Immunol 2023; 14:1308143. [PMID: 38187402 PMCID: PMC10768003 DOI: 10.3389/fimmu.2023.1308143] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Background Ovarian cancer (OC) is one of the malignant tumors that poses a serious threat to women's health. Natural killer (NK) cells are an integral part of the immune system and have the ability to kill tumor cells directly or participate indirectly in the anti-tumor immune response. In recent years, NK cell-based immunotherapy for OC has shown remarkable potential. However, its mechanisms and effects remain unclear when compared to standard treatment. Methods To explore the value of NK cell-based immunotherapy in the treatment of OC, we conducted a literature review. In comparison to standard treatment, our focus was primarily on the current anti-tumor mechanisms, the clinical effect of NK cells against OC, factors affecting the structure and function of NK cells, and strategies to enhance the effectiveness of NK cells. Results We found that NK cells exert their therapeutic effects in OC through mechanisms such as antibody-dependent cell cytotoxicity, perforin release, and granule enzyme secretion. They also secrete IFN-γ and TNF-α or engage in Fas/FasL and TRAIL/TRAILR pathways, mediating the death of OC cells. In clinical trials, the majority of patients experienced disease stability with mild side effects after receiving NK cell-based immunotherapy, but there is still a lack of high-quality research evidence regarding its clinical effectiveness. OC and prior experience with standard treatments have an effect on NK cells, and it may be considered to maximize NK cell effects through the modulation of the tumor microenvironment or combination with other therapies. Conclusions In this review, we have summarized the current evidence of NK cell applications in the treatment of OC. Furthermore, factors and strategies that influence and enhance the role of NK cell immunotherapy are discussed.
Collapse
Affiliation(s)
- Zhongru Fan
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Dongyu Han
- Department of Obstetrics and Gynecology, Suzhou Hospital, Affiliated Hospital of Meddical School, Nanjing University, Suzhou, China
| | - Xin Fan
- Department of Radiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Lin Zhao
- Department of Obstetrics and Gynecology, Suzhou Hospital, Affiliated Hospital of Meddical School, Nanjing University, Suzhou, China
| |
Collapse
|
19
|
Du Y, Pollok KE, Shen J. Unlocking Glioblastoma Secrets: Natural Killer Cell Therapy against Cancer Stem Cells. Cancers (Basel) 2023; 15:5836. [PMID: 38136381 PMCID: PMC10741423 DOI: 10.3390/cancers15245836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/27/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Glioblastoma (GBM) represents a paramount challenge as the most formidable primary brain tumor characterized by its rapid growth, aggressive invasiveness, and remarkable heterogeneity, collectively impeding effective therapeutic interventions. The cancer stem cells within GBM, GBM stem cells (GSCs), hold pivotal significance in fueling tumor advancement, therapeutic refractoriness, and relapse. Given their unique attributes encompassing self-renewal, multipotent differentiation potential, and intricate interplay with the tumor microenvironment, targeting GSCs emerges as a critical strategy for innovative GBM treatments. Natural killer (NK) cells, innate immune effectors recognized for their capacity to selectively detect and eliminate malignancies without the need for prior sensitization, offer substantial therapeutic potential. Harnessing the inherent capabilities of NK cells can not only directly engage tumor cells but also augment broader immune responses. Encouraging outcomes from clinical investigations underscore NK cells as a potentially effective modality for cancer therapy. Consequently, NK cell-based approaches hold promise for effectively targeting GSCs, thereby presenting an avenue to enhance treatment outcomes for GBM patients. This review outlines GBM's intricate landscape, therapeutic challenges, GSC-related dynamics, and elucidates the potential of NK cell as an immunotherapeutic strategy directed towards GSCs.
Collapse
Affiliation(s)
- Yuanning Du
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA;
| | - Karen E. Pollok
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Department of Pediatrics, Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | - Jia Shen
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA;
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| |
Collapse
|
20
|
McMahon RA, D'Souza C, Neeson PJ, Siva S. Innate immunity: Looking beyond T-cells in radiation and immunotherapy combinations. Neoplasia 2023; 46:100940. [PMID: 37913654 PMCID: PMC10637988 DOI: 10.1016/j.neo.2023.100940] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 11/03/2023]
Abstract
Radiation therapy is an established and effective anti-cancer treatment modality. Extensive pre-clinical experimentation has demonstrated that the pro-inflammatory properties of irradiation may be synergistic with checkpoint immunotherapy. Radiation induces double-stranded DNA breaks (dsDNA). Sensing of the dsDNA activates the cGAS/STING pathway, producing Type 1 interferons essential to recruiting antigen-presenting cells (APCs). Radiation promotes cytotoxic CD8 T-cell recruitment by releasing tumour-associated antigens captured and cross-presented by surveying antigen-presenting cells. Radiation-induced vascular normalisation may further promote T-cell trafficking and drug delivery. Radiation is also immunosuppressive. Recruitment of regulatory T cells (Tregs) and innate cells such as myeloid-derived suppressive cells (m-MDSCs) all counteract the immunostimulatory properties of radiation. Many innate immune cell types operate at the interface of the adaptive immune response. Innate immune cells, such as m-MDSCs, can exert their immunosuppressive effects by expressing immune checkpoints such as PD-L1, further highlighting the potential of combined radiation and checkpoint immunotherapy. Several early-phase clinical studies investigating the combination of radiation and immunotherapy have been disappointing. A greater appreciation of radiotherapy's impact on the innate immune system is essential to optimise radioimmunotherapy combinations. This review will summarise the impact of radiotherapy on crucial cells of the innate immune system and vital immunosuppressive cytokines.
Collapse
Affiliation(s)
- R A McMahon
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia.
| | - C D'Souza
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia; Cancer Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - P J Neeson
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia; Cancer Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - S Siva
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia
| |
Collapse
|
21
|
Saini S, Gangwar A, Sharma R. Harnessing host-pathogen interactions for innovative drug discovery and host-directed therapeutics to tackle tuberculosis. Microbiol Res 2023; 275:127466. [PMID: 37531813 DOI: 10.1016/j.micres.2023.127466] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/04/2023]
Abstract
Tuberculosis (TB) is a highly contagious bacterial infection caused by Mycobacterium tuberculosis (Mtb), which has been ranked as the second leading cause of death worldwide from a single infectious agent. As an intracellular pathogen, Mtb has well adapted to the phagocytic host microenvironment, influencing diverse host processes such as gene expression, trafficking, metabolism, and signaling pathways of the host to its advantage. These responses are the result of dynamic interactions of the bacteria with the host cell signaling pathways, whereby the bacteria attenuate the host cellular processes for their survival. Specific host genes and the mechanisms involved in the entry and subsequent stabilization of M. tuberculosis intracellularly have been identified in various genetic and chemical screens recently. The present understanding of the co-evolution of Mtb and macrophage system presented us the new possibilities for exploring host-directed therapeutics (HDT). Here, we discuss the host-pathogen interaction for Mtb, including the pathways adapted by Mtb to escape immunity. The review sheds light on different host-directed therapies (HDTs) such as repurposed drugs and vitamins, along with their targets such as granuloma, autophagy, extracellular matrix, lipids, and cytokines, among others. The article also examines the available clinical data on these drug molecules. In conclusion, the review presents a perspective on the current knowledge in the field of HDTs and the need for additional research to overcome the challenges associated HDTs.
Collapse
Affiliation(s)
- Sapna Saini
- Infectious Diseases Division, CSIR, Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anjali Gangwar
- Infectious Diseases Division, CSIR, Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rashmi Sharma
- Infectious Diseases Division, CSIR, Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
22
|
Sheikh K, Memon KN, Usman H, Abdel-Maksoud MA, Ullah S, Almanaa TN, Chaudhary A, Jamil M, Gill OBQ, Yar MA, Hussein AM, Zakri AM. Identification of useful biomolecular markers in kidney renal clear cell carcinoma: an in silico and in vitro analysis-based study. Am J Transl Res 2023; 15:5574-5593. [PMID: 37854221 PMCID: PMC10579006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 08/28/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND Kidney renal clear cell carcinoma (KIRC) is the most prevalent type of renal cell carcinoma (RCC), with a high incidence and mortality rate. There is a lack of sensitive biomarkers. Therefore, the discovery of accurate biomarkers for KIRC patients is critical to improve prognosis. METHODS We determined hub genes and their associated pathways involved in the pathogenesis of KIRC from the GSE66272 dataset consisting of KIRC (n = 26) and corresponding control (n = 26) samples and later validated the expression and methylation level of the identified hub genes on The Cancer Genomic Atlas (TCGA) datasets and Human RCC 786-O and normal HK-2 cell lines through RNA sequencing (RNA-seq), Reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and targeted bisulfite sequencing (bisulfite-seq) analyses. RESULTS The identified up-regulated four hub genes include TYROBP (Transmembrane Immune Signaling Adaptor TYROBP), PTPRC (Protein tyrosine phosphatase, receptor type, C), LCP2 (Lymphocyte cytosolic protein 2), and ITGB2 (Integrin Subunit Beta 2). Moreover, the higher expression of TYROBP, PTPRC, LCP2, and ITGB2 in KIRC patients insignificantly correlates with a poor prognosis in KIRC patients. In addition, hub genes were involved in the "Fc epsilon RI signaling pathway, asthma, natural cell killer mediated cytotoxicity, T cell receptor signaling pathway, primary immunodeficiency, Fc gamma R-mediated phagocytosis, malaria, leukocyte transendothelial migration, and legionellosis" pathways and associated with the infiltration level of CD8+ T, CD4+ T, and macrophage cells. CONCLUSION Our integrated in silico and in vitro analysis identified important hub genes (TYROBP, PTPRC, LCP2, and ITGB2) involved in the pathogenesis of KIRC as possible diagnostic biomarkers.
Collapse
Affiliation(s)
- Khalida Sheikh
- Liaquat University of Medical and Health Sciences (LUMHS)Jamshoro 76090, Pakistan
| | | | - Humera Usman
- Fazaia Medical College, Air UniversityIslamabad 44000, Pakistan
| | | | | | | | - Aqsa Chaudhary
- Department of Biochemistry, University of Central PunjabLahore, Pakistan
| | - Muhammad Jamil
- PARC Arid Zone Research CenterDera Ismail Khan 29050, Pakistan
| | | | - Muhammad Ahmed Yar
- Mufti Mehmood Memorial Teaching HospitalDera Ismail Khan 29050, KPK, Pakistan
| | - Ahmed M Hussein
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of ViennaVienna 1090, Austria
| | - Adel M Zakri
- Plant Production Department, College of Food and Agricultural Sciences, King Saud UniversityRiyadh 11451, Saudi Arabia
| |
Collapse
|
23
|
Hwang TL, Chang CH. Oridonin enhances cytotoxic activity of natural killer cells against lung cancer. Int Immunopharmacol 2023; 122:110669. [PMID: 37480753 DOI: 10.1016/j.intimp.2023.110669] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/07/2023] [Accepted: 07/16/2023] [Indexed: 07/24/2023]
Abstract
BACKGROUND Oridonin is a Chinese herbal medicine exhibiting anti-tumor properties; however, its immune modulation capacity has yet to be elucidated. Our objective in this study was to determine whether oridonin enhances the anti-tumor activity of natural killer (NK) cells against lung cancer cells. METHODS LDH-releasing assays were used to investigate the effects of oridonin on NK-92MI cell activity against lung cancer cells. Flow cytometry and real-time PCR were used to examine the effects of oridonin on degranulation markers, cytotoxic factors, activating receptors on NK-92MI cells, and ligands in lung cancer cells. Western blot analysis provided insight into the mechanisms underlying the observed effects. RESULTS Oridonin enhanced the cytotoxic effects of NK-92MI cells against A549 lung cancer cells. This effect involved upregulating the expression of the degranulation marker CD107a and IFN-γ as well as activating receptors on NK cells and their ligand MICA/B. Oridonin also inhibited STAT3 phosphorylation in A549 cells and NK-92MI cells. A lung cancer mouse model confirmed the anti-tumor effects of oridonin and NK-92MI cells, wherein both treatments alone suppressed tumor growth. Oridonin was also shown to have a synergistic effect on the anti-tumor activity of NK-92MI cells. CONCLUSIONS The ability of oridonin to enhance the cytotoxic effects of NK cells indicates its potential as a novel therapeutic agent for the treatment of lung cancer.
Collapse
Affiliation(s)
- Tsong-Long Hwang
- Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan; Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, 243, Taiwan.
| | - Chuan-Hsin Chang
- Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan.
| |
Collapse
|
24
|
Rahmati A, Bigam S, Elahi S. Galectin-9 promotes natural killer cells activity via interaction with CD44. Front Immunol 2023; 14:1131379. [PMID: 37006235 PMCID: PMC10060867 DOI: 10.3389/fimmu.2023.1131379] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
Natural killer (NK) cells are a potent innate source of cytokines and cytoplasmic granules. Their effector functions are tightly synchronized by the balance between the stimulatory and inhibitory receptors. Here, we quantified the proportion of NK cells and the surface presence of Galectin-9 (Gal-9) from the bone marrow, blood, liver, spleen, and lungs of adult and neonatal mice. We also examined the effector functions of Gal-9+NK cells compared with their Gal-9- counterparts. Our results revealed that Gal-9+NK cells are more abundant in tissues, in particular, in the liver than in the blood and bone marrow. We found Gal-9 presence was associated with enhanced cytotoxic effector molecules granzyme B (GzmB) and perforin expression. Likewise, Gal-9 expressing NK cells displayed greater IFN-γ and TNF-α expression than their negative counterparts under hemostatic circumstances. Notably, the expansion of Gal-9+NK cells in the spleen of mice infected with E. coli implies that Gal-9+NK cells may provide a protective role against infection. Similarly, we found the expansion of Gal-9+NK cells in the spleen and tumor tissues of melanoma B16-F10 mice. Mechanistically, our results revealed the interaction of Gal-9 with CD44 as noted by their co-expression/co-localization. Subsequently, this interaction resulted in enhanced expression of Phospho-LCK, ERK, Akt, MAPK, and mTOR in NK cells. Moreover, we found Gal-9+NK cells exhibited an activated phenotype as evidenced by increased CD69, CD25, and Sca-1 but reduced KLRG1 expression. Likewise, we found Gal-9 preferentially interacts with CD44high in human NK cells. Despite this interaction, we noted a dichotomy in terms of effector functions in NK cells from COVID-19 patients. We observed that the presence of Gal-9 on NK cells resulted in a greater IFN-γ expression without any changes in cytolytic molecule expression in these patients. These observations suggest differences in Gal-9+NK cell effector functions between mice and humans that should be considered in different physiological and pathological conditions. Therefore, our results highlight the important role of Gal-9 via CD44 in NK cell activation, which suggests Gal-9 is a potential new avenue for the development of therapeutic approaches to modulate NK cell effector functions.
Collapse
Affiliation(s)
- Amirhossein Rahmati
- School of Dentistry, Division of Foundational Sciences, University of Alberta, Edmonton, AB, Canada
| | - Steven Bigam
- School of Dentistry, Division of Foundational Sciences, University of Alberta, Edmonton, AB, Canada
| | - Shokrollah Elahi
- School of Dentistry, Division of Foundational Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
- Li Ka Shing Institute of Virology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Shokrollah Elahi,
| |
Collapse
|
25
|
Wei X, Yang X. The central role of natural killer cells in preeclampsia. Front Immunol 2023; 14:1009867. [PMID: 36865565 PMCID: PMC9972679 DOI: 10.3389/fimmu.2023.1009867] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Preeclampsia (PE) is a disease that is unique to pregnancy and affects multiple systems. It can lead to maternal and perinatal mortality. The precise etiology of PE is unclear. Patients with PE may have systemic or local immune abnormalities. A group of researchers has proposed that the immune communication between the fetus and mother is primarily moderated by natural killer (NK) cells as opposed to T cells, since NK cells are the most abundant immune cells in the uterus. This review examines the immunological roles of NK cells in the pathogenesis of PE. Our aim is to provide obstetricians with a comprehensive and updated research progress report on NK cells in PE patients. It has been reported that decidual NK (dNK) cells contribute to the process of uterine spiral artery remodeling and can modulate trophoblast invasion. Additionally, dNK cells can stimulate fetal growth and regulate delivery. It appears that the count or proportion of circulating NK cells is elevated in patients with or at risk for PE. Changes in the number or function of dNK cells may be the cause of PE. The Th1/Th2 equilibrium in PE has gradually shifted to an NK1/NK2 equilibrium based on cytokine production. An improper combination of killer cell immunoglobulin-like receptor (KIR) and human leukocyte antigen (HLA)-C may lead to insufficient activation of dNK cells, thereby causing PE. In the etiology of PE, NK cells appear to exert a central effect in both peripheral blood and the maternal-fetal interface. To maintain immune equilibrium both locally and systemically, it is necessary to take therapeutic measures directed at NK cells.
Collapse
Affiliation(s)
- Xiaoqi Wei
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, China
| | - Xiuhua Yang
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
26
|
Zhang X, Jiang Y, Li S, Bian D, Liu M, Kong M, Chen Y, Duan Z, Zheng S. Direct-acting Antiviral-induced Transient Recovery of NK Cells in Early-stage Treatment of Chronic Hepatitis C Patients. J Clin Transl Hepatol 2022; 10:1117-1124. [PMID: 36381106 PMCID: PMC9634778 DOI: 10.14218/jcth.2021.00427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/30/2021] [Accepted: 12/27/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND AIMS The rapid clearance of hepatitis C virus induced by direct-acting antivirals (DAAs) affects natural killer (NK) cells, but the reported results are not consistent, and the relative mechanism was unclear. This study focused on the dynamic changes of NK cells during and after DAA treatment and analyzed the reasons. METHODS Peripheral blood from 35 chronic hepatitis C patients who were treated with DAAs were collected at baseline and weeks 1, 2, 4, 12, and post-treatment week-12. The frequency, subset, and phenotype of NK cells were assayed by flow cytometry. Lactate dehydrogenase assays were used to evaluate the cytotoxicity of NK cells. Cytokine concentrations were measured with Luminex kits. RESULTS All patients achieved a sustained viral response (SVR), and the NK cell frequencies were not changed significantly during DAA therapy. However, the cytotoxicity of NK cells recovered significantly early in week 1, and then continuously decreased below normal levels. The changes of genotypes including NKp30+, NKp46+, and NKG2A+ NK cells were parallel to NK function. The subset of CD56dim NK cells continuously increased and did not return to normal even at 12 weeks after treatment. Interleukin (IL)-2, IL10, IL15, interferon-gamma, and tumor necrosis factor-alpha all increased after week 4, peaked at the end of therapy, and then exhibited varying degrees of reduction with time. CONCLUSIONS DAA treatment led to transient functional recovery of NK cells in the early stage of treatment, and then continuously decreased to below normal levels. Alterations of NK subsets, phenotypes, and the microenvironment may be involved in the changes.
Collapse
Affiliation(s)
- Xiaohui Zhang
- The Fourth Department of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment & Research, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Yingying Jiang
- Department of Gastroenterology and Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Shaobin Li
- School of Energy and Power Engineering,Beihang University, Beijing, China
| | - Dandan Bian
- Department of Infectious Diseases, Electric Power Teaching Hospital, Capital Medical University, Beijing, China
| | - Mei Liu
- Oncology Department, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Ming Kong
- The Fourth Department of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment & Research, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Yu Chen
- The Fourth Department of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment & Research, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Zhongping Duan
- The Fourth Department of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment & Research, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Sujun Zheng
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment & Research, Beijing YouAn Hospital, Capital Medical University, Beijing, China
- The First Department of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
27
|
Lv XQ, Zhang KB, Guo X, Pei L, Li F. Higher TYROBP and lower SOX6 as predictive biomarkers for poor prognosis of clear cell renal cell carcinoma: A pilot study. Medicine (Baltimore) 2022; 101:e30658. [PMID: 36595751 PMCID: PMC9794311 DOI: 10.1097/md.0000000000030658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Clear cell renal carcinoma (ccRCC) is the most common subtype of renal cancer, accounting for approximately 75% of all histological types of renal cancer, and is the leading cause of death from renal cancer. However, the molecular mechanism of tyrosine kinase binding protein (TYROBP) and sex-determining region Y Box-6 (SOX6) in the ccRCC was not precise. METHODS Bioinformatics analysis was performed to explore the hub role of TYROBP and SOX6 on the ccRCC. A total of 6 patients with clear cell renal cell carcinoma (ccRCC) were recruited. HE staining was performed to observe the pathology result of ccRCC. Immunohistochemistry and Immunofluorescence assay was made to detect the protein expression of TYROBP. Total RNA was extracted using TRIzol to examine the mRNA expression of TYROBP via the Real time quantitative polymerase chain reaction. The strong correlation between the expression of TYROBP and the survival time of ccRCC patients was performed by the BP neural network and support vector machine. RESULTS Compared with the control group, the expression of SOX6 was downregulated in the samples with ccRCC. However, the expression of TYROBP was higher in the samples with ccRCC than in the control group. Compared with the patients with high SOX6 expression, the patients with low SOX6 expression have a poor survival prognosis (HR=0.39, P < .05). However, the patients with high TYROBP expression have a shorter survival time than the patients with low TYROBP expression (HR=1.66, P < .05). The genes related with TYROBP and SOX6 are mainly enriched in the regulation of cell activation, leukocyte activation, negative regulation of cell activation, myeloid leukocyte activation, positive regulation of response to external stimulus, immune response-regulating signaling pathway. The interaction between TYROBP, SOX6, and kidney neoplasms was drawn, and the inference score of TYROBP and SOX6 on the kidney neoplasms was high. CONCLUSION In conclusion, TYROBP is highly expressed in renal clear cell carcinoma, and when this molecule is highly expressed, the survival prognosis of renal carcinoma is poor. TYROBP and SOX6 may be potential targets for diagnosing and treating renal clear cell carcinoma.
Collapse
Affiliation(s)
- Xian-Qiang Lv
- Department of Urology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Kai-bo Zhang
- Department of plastic surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Xu Guo
- Lab of Gambridge Analytica, Heping Road, Shijiazhuang, Hebei Province, China
| | - Long Pei
- Department of Urology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Feng Li
- Department of Urology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
- * Correspondence: Feng Li, Department of Urology, The Fourth Hospital of Hebei Medical University, No.12 Jiankang Road Shijiazhuang, Hebei Province 050011, China (e-mail: )
| |
Collapse
|
28
|
Donor natural killer cells trigger production of β-2-microglobulin to enhance post-bone marrow transplant immunity. Blood 2022; 140:2323-2334. [PMID: 35984965 DOI: 10.1182/blood.2021015297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 07/14/2022] [Indexed: 11/20/2022] Open
Abstract
Allogeneic hematopoietic transplantation is a powerful treatment for hematologic malignancies. Posttransplant immune incompetence exposes patients to disease relapse and infections. We previously demonstrated that donor alloreactive natural killer (NK) cells ablate recipient hematopoietic targets, including leukemia. Here, in murine models, we show that infusion of donor alloreactive NK cells triggers recipient dendritic cells (DCs) to synthesize β-2-microglobulin (B2M) that elicits the release of c-KIT ligand and interleukin-7 that greatly accelerate posttransplant immune reconstitution. An identical chain of events was reproduced by infusing supernatants of alloreactive NK/DC cocultures. Similarly, human alloreactive NK cells triggered human DCs to synthesize B2M that induced interleukin-7 production by thymic epithelial cells and thereby supported thymocyte cellularity in vitro. Chromatography fractionation of murine and human alloreactive NK/DC coculture supernatants identified a protein with molecular weight and isoelectric point of B2M, and mass spectrometry identified amino acid sequences specific of B2M. Anti-B2M antibody depletion of NK/DC coculture supernatants abrogated their immune-rebuilding effect. B2M knock-out mice were unable to undergo accelerated immune reconstitution, but infusion of (wild-type) NK/DC coculture supernatants restored their ability to undergo accelerated immune reconstitution. Similarly, silencing the B2M gene in human DCs, before coculture with alloreactive NK cells, prevented the increase in thymocyte cellularity in vitro. Finally, human recombinant B2M increased thymocyte cellularity in a thymic epithelial cells/thymocyte culture system. Our studies uncover a novel therapeutic principle for treating posttransplant immune incompetence and suggest that, upon its translation to the clinic, patients may benefit from adoptive transfer of large numbers of cytokine-activated, ex vivo-expanded donor alloreactive NK cells.
Collapse
|
29
|
Restricted Recruitment of NK Cells with Impaired Function Is Caused by HPV-Driven Immunosuppressive Microenvironment of Papillomas in Aggressive Juvenile-Onset Recurrent Respiratory Papillomatosis Patients. J Virol 2022; 96:e0094622. [PMID: 36154611 DOI: 10.1128/jvi.00946-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Laryngopharynx epithelium neoplasia induced by HPV6/11 infection in juvenile-onset recurrent respiratory papillomatosis (JO-RRP) causes a great health issue characteristic of frequent relapse and aggressive disease progression. Local cell-mediated immunity shaped by the recruitment and activation of cytotoxic effector cells is critical for viral clearance. In this study, we found that NK cells in the papillomas of aggressive JO-RRP patients, in contrast to massive infiltrated T cells, were scarce in number and impaired in activation and cytotoxicity as they were in peripheral blood. Data from cell infiltration analysis indicated that the migration of NK cell to papilloma was restricted in aggressive JO-RRP patients. Further study showed that the skewed chemokine expression in the papillomas and elevated ICAM-1 expression in hyperplastic epithelia cells favored the T cell but not NK cell recruitment in aggressive JO-RRP patients. In parallel to the increased CD3+ T cells, we observed a dramatical increase in Tregs and Treg-promoting cytokines such as IL-4, IL-10 and TGFβ in papillomas of aggressive JO-RRP patients. Our study suggested that likely initialized by the intrinsic change in neoplastic epithelial cells with persistent HPV infection, the aggressive papillomas built an entry barrier for NK cell infiltration and formed an immunosuppressive clump to fend off the immune attack from intra-papillomas NK cells. IMPORTANCE Frequent relapse and aggressive disease progression of juvenile-onset recurrent respiratory papillomatosis (JO-RRP) pose a great challenge to the complete remission of HPV 6/11 related laryngeal neoplasia. Local immune responses in papillomas are more relevant to the disease control considering the locale infected restriction of HPV virus in epitheliums. In our study, the restricted NK cell number and reduced expression of activating NKp30 receptor suggested one possible mechanism underlying impaired NK cell defense ability in aggressive JO-RRP papillomas. Meanwhile, the negative impact of HPV persistent infection on NK cell number and function represented yet another example of a chronic pathogen subverting NK cell behavior, affirming a potentially important role for NK cells in viral containment. Further, the skewed chemokine/cytokine expression in the papillomas and the elevated adhesion molecules expression in hyperplastic epithelia cells provided important clues for understanding blocked infiltration and antiviral dysfunction of NK cells in papilloma.
Collapse
|
30
|
Li X, Wichai N, Wang J, Liu X, Yan H, Wang Y, Luo M, Zhou S, Wang K, Li L, Miao L. Regulation of innate and adaptive immunity using herbal medicine: benefits for the COVID-19 vaccination. ACUPUNCTURE AND HERBAL MEDICINE 2022; 2:196-206. [PMID: 37808346 PMCID: PMC9746255 DOI: 10.1097/hm9.0000000000000046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 09/06/2022] [Indexed: 08/18/2023]
Abstract
Vaccination is a major achievement that has become an effective prevention strategy against infectious diseases and active control of emerging pathogens worldwide. In response to the coronavirus disease 2019 (COVID-19) pandemic, several diverse vaccines against severe acute respiratory syndrome coronavirus 2 have been developed and deployed for use in a large number of individuals, and have been reported to protect against symptomatic COVID-19 cases and deaths. However, the application of vaccines has a series of limitations, including protective failure for variants of concern, unavailability of individuals due to immune deficiency, and the disappearance of immune protection for increasing infections in vaccinated individuals. These aspects raise the question of how to modulate the immune system that contributes to the COVID-19 vaccine protective effects. Herbal medicines are widely used for their immune regulatory abilities in clinics. More attractively, herbal medicines have been well accepted for their positive role in the COVID-19 prevention and suppression through regulation of the immune system. This review presents a brief overview of the strategy of COVID-19 vaccination and the response of the immune system to vaccines, the regulatory effects and mechanisms of herbal medicine in immune-related macrophages, natural killer cells, dendritic cells, and lymphocytes T and B cells, and how they help vaccines work. Later in the article, the potential role and application of herbal medicines in the most recent COVID-19 vaccination are discussed. This article provides new insights into herbal medicines as promising alternative supplements that may benefit from COVID-19 vaccination. Graphical abstract http://links.lww.com/AHM/A31.
Collapse
Affiliation(s)
- Xuan Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Nuttapong Wichai
- Faculty of Pharmacy, Mahasarakham University, Mahasarakham, Thailand
| | - Jiabao Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiuping Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huimin Yan
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yu Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Mingchi Luo
- Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shengyuan Zhou
- Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Kai Wang
- Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Miao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
31
|
Lopez KJ, Cross-Najafi AA, Farag K, Obando B, Thadasina D, Isidan A, Park Y, Zhang W, Ekser B, Li P. Strategies to induce natural killer cell tolerance in xenotransplantation. Front Immunol 2022; 13:941880. [PMID: 36072599 PMCID: PMC9441937 DOI: 10.3389/fimmu.2022.941880] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/28/2022] [Indexed: 11/23/2022] Open
Abstract
Eliminating major xenoantigens in pig cells has drastically reduced human antibody-mediated hyperacute xenograft rejection (HXR). Despite these advancements, acute xenograft rejection (AXR) remains one of the major obstacles to clinical xenotransplantation, mediated by innate immune cells, including macrophages, neutrophils, and natural killer (NK) cells. NK cells play an 'effector' role by releasing cytotoxicity granules against xenogeneic cells and an 'affecter' role on other immune cells through cytokine secretion. We highlight the key receptor-ligand interactions that determine the NK cell response to target cells, focusing on the regulation of NK cell activating receptor (NKG2D, DNAM1) and inhibitory receptor (KIR2DL1-4, NKG2A, and LIR-1) signaling pathways. Inhibition of NK cell activity may protect xenografts from cytotoxicity. Recent successful approaches to reducing NK cell-mediated HXR and AXR are reviewed, including genetic modifications of porcine xenografts aimed at improving pig-to-human compatibility. Future directions to promote xenograft acceptance are discussed, including NK cell tolerance in pregnancy and NK cell evasion in viral infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ping Li
- *Correspondence: Ping Li, ; Burcin Ekser,
| |
Collapse
|
32
|
Focaccetti C, Benvenuto M, Pighi C, Vitelli A, Napolitano F, Cotugno N, Fruci D, Palma P, Rossi P, Bei R, Cifaldi L. DNAM-1-chimeric receptor-engineered NK cells, combined with Nutlin-3a, more effectively fight neuroblastoma cells in vitro: a proof-of-concept study. Front Immunol 2022; 13:886319. [PMID: 35967339 PMCID: PMC9367496 DOI: 10.3389/fimmu.2022.886319] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 07/01/2022] [Indexed: 11/16/2022] Open
Abstract
Adoptive transfer of engineered NK cells, one of clinical approaches to fight cancer, is gaining great interest in the last decade. However, the development of new strategies is needed to improve clinical efficacy and safety of NK cell-based immunotherapy. NK cell-mediated recognition and lysis of tumor cells are strictly dependent on the expression of ligands for NK cell-activating receptors NKG2D and DNAM-1 on tumor cells. Of note, the PVR/CD155 and Nectin-2/CD112 ligands for DNAM-1 are expressed primarily on solid tumor cells and poorly expressed in normal tissue cells. Here, we generated human NK cells expressing either the full length DNAM-1 receptor or three different DNAM-1-based chimeric receptor that provide the expression of DNAM-1 fused to a costimulatory molecule such as 2B4 and CD3ζ chain. Upon transfection into primary human NK cells isolated from healthy donors, we evaluated the surface expression of DNAM-1 and, as a functional readout, we assessed the extent of degranulation, cytotoxicity and the production of IFNγ and TNFα in response to human leukemic K562 cell line. In addition, we explored the effect of Nutlin-3a, a MDM2-targeting drug able of restoring p53 functions and known to have an immunomodulatory effect, on the degranulation of DNAM-1-engineered NK cells in response to human neuroblastoma (NB) LA-N-5 and SMS-KCNR cell lines. By comparing NK cells transfected with four different plasmid vectors and through blocking experiments, DNAM-1-CD3ζ-engineered NK cells showed the strongest response. Furthermore, both LA-N-5 and SMS-KCNR cells pretreated with Nutlin-3a were significantly more susceptible to DNAM-1-engineered NK cells than NK cells transfected with the empty vector. Our results provide a proof-of-concept suggesting that the combined use of DNAM-1-chimeric receptor-engineered NK cells and Nutlin-3a may represent a novel therapeutic approach for the treatment of solid tumors, such as NB, carrying dysfunctional p53.
Collapse
Affiliation(s)
- Chiara Focaccetti
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Monica Benvenuto
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Rome, Italy
- Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - Chiara Pighi
- Research Unit of Clinical Immunology and Vaccinology, Dipartimento Pediatrico Universitario Ospedaliero (DPUO), Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | | | | | - Nicola Cotugno
- Research Unit of Clinical Immunology and Vaccinology, Dipartimento Pediatrico Universitario Ospedaliero (DPUO), Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Chair of Pediatrics, Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Doriana Fruci
- Department of Paediatric Haematology/Oncology and of Cell and Gene Therapy, Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Paolo Palma
- Research Unit of Clinical Immunology and Vaccinology, Dipartimento Pediatrico Universitario Ospedaliero (DPUO), Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Chair of Pediatrics, Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Paolo Rossi
- Chair of Pediatrics, Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
- Academic Department of Pediatrics (DPUO), Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Loredana Cifaldi
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Rome, Italy
- Academic Department of Pediatrics (DPUO), Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- *Correspondence: Loredana Cifaldi,
| |
Collapse
|
33
|
Yu T, Yu SK, Xiang Y, Lu KH, Sun M. Revolution of CAR Engineering For Next-Generation Immunotherapy In Solid Tumors. Front Immunol 2022; 13:936496. [PMID: 35903099 PMCID: PMC9315443 DOI: 10.3389/fimmu.2022.936496] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/16/2022] [Indexed: 01/01/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T cells have enormous potentials for clinical therapies. The CAR-T therapy has been approved for treating hematological malignancies. However, their application is limited in solid tumors owing to antigen loss and mutation, physical barriers, and an immunosuppressive tumor microenvironment. To overcome the challenges of CAR-T, increasing efforts are put into developing CAR-T to expand its applied ranges. Varied receptors are utilized for recognizing tumor-associated antigens and relieving immunosuppression. Emerging co-stimulatory signaling is employed for CAR-T activation. Furthermore, other immune cells such as NK cells and macrophages have manifested potential for delivering CAR. Hence, we collected and summarized the last advancements of CAR engineering from three aspects, namely, the ectodomains, endogenous domains, and immune cells, aiming to inspire the design of next-generation adoptive immunotherapy for treating solid tumors.
Collapse
Affiliation(s)
- Tao Yu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shao-kun Yu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Xiang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kai-Hua Lu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Kai-Hua Lu, ; Ming Sun,
| | - Ming Sun
- Suzhou Cancer Center Core Laboratory, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- *Correspondence: Kai-Hua Lu, ; Ming Sun,
| |
Collapse
|
34
|
Boyd-Gibbins N, Karagiannis P, Hwang DW, Kim SI. iPSCs in NK Cell Manufacturing and NKEV Development. Front Immunol 2022; 13:890894. [PMID: 35874677 PMCID: PMC9305199 DOI: 10.3389/fimmu.2022.890894] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 06/03/2022] [Indexed: 11/27/2022] Open
Abstract
Natural killer (NK) cell immunotherapies for cancer can complement existing T cell therapies while benefiting from advancements already made in the immunotherapy field. For NK cell manufacturing, induced pluripotent stem cells (iPSCs) offer advantages including eliminating donor variation and providing an ideal platform for genome engineering. At the same time, extracellular vesicles (EVs) have become a major research interest, and purified NK cell extracellular vesicles (NKEVs) have been shown to reproduce the key functions of their parent NK cells. NKEVs have the potential to be developed into a standalone therapeutic with reduced complexity and immunogenicity compared to cell therapies. This review explores the role iPSC technology can play in both NK cell manufacturing and NKEV development.
Collapse
Affiliation(s)
| | - Peter Karagiannis
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Do Won Hwang
- Research and Development Center, THERABEST Co., Ltd., Seoul, South Korea
| | - Shin-Il Kim
- THERABEST Japan, Inc., Kobe, Japan
- Research and Development Center, THERABEST Co., Ltd., Seoul, South Korea
| |
Collapse
|
35
|
Garcinuño S, Gil-Etayo FJ, Mancebo E, López-Nevado M, Lalueza A, Díaz-Simón R, Pleguezuelo DE, Serrano M, Cabrera-Marante O, Allende LM, Paz-Artal E, Serrano A. Effective Natural Killer Cell Degranulation Is an Essential Key in COVID-19 Evolution. Int J Mol Sci 2022; 23:ijms23126577. [PMID: 35743021 PMCID: PMC9224310 DOI: 10.3390/ijms23126577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 02/05/2023] Open
Abstract
NK degranulation plays an important role in the cytotoxic activity of innate immunity in the clearance of intracellular infections and is an important factor in the outcome of the disease. This work has studied NK degranulation and innate immunological profiles and functionalities in COVID-19 patients and its association with the severity of the disease. A prospective observational study with 99 COVID-19 patients was conducted. Patients were grouped according to hospital requirements and severity. Innate immune cell subpopulations and functionalities were analyzed. The profile and functionality of innate immune cells differ between healthy controls and severe patients; CD56dim NK cells increased and MAIT cells and NK degranulation rates decreased in the COVID-19 subjects. Higher degranulation rates were observed in the non-severe patients and in the healthy controls compared to the severe patients. Benign forms of the disease had a higher granzymeA/granzymeB ratio than complex forms. In a multivariate analysis, the degranulation capacity resulted in a protective factor against severe forms of the disease (OR: 0.86), whereas the permanent expression of NKG2D in NKT cells was an independent risk factor (OR: 3.81; AUC: 0.84). In conclusion, a prompt and efficient degranulation functionality in the early stages of infection could be used as a tool to identify patients who will have a better evolution.
Collapse
Affiliation(s)
- Sara Garcinuño
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
| | - Francisco Javier Gil-Etayo
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Esther Mancebo
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Marta López-Nevado
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
| | - Antonio Lalueza
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain;
| | - Raquel Díaz-Simón
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain;
| | - Daniel Enrique Pleguezuelo
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Manuel Serrano
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Oscar Cabrera-Marante
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Luis M. Allende
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
- Department of Immunology, Ophthalmology and Otorhinolaryngology, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Estela Paz-Artal
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
- Department of Immunology, Ophthalmology and Otorhinolaryngology, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Antonio Serrano
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.G.); (F.J.G.-E.); (E.M.); (M.L.-N.); (A.L.); (D.E.P.); (M.S.); (O.C.-M.); (L.M.A.); (E.P.-A.)
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
- Biomedical Research Centre Network for Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-652-085-293
| |
Collapse
|
36
|
Sönmez C, Wölfer J, Holling M, Brokinkel B, Stummer W, Wiendl H, Thomas C, Schulte-Mecklenbeck A, Grauer OM. Blockade of inhibitory killer cell immunoglobulin-like receptors and IL-2 triggering reverses the functional hypoactivity of tumor-derived NK-cells in glioblastomas. Sci Rep 2022; 12:6769. [PMID: 35474089 PMCID: PMC9042843 DOI: 10.1038/s41598-022-10680-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/11/2022] [Indexed: 11/09/2022] Open
Abstract
Killer cell immunoglobulin-like receptors (KIRs) comprise a group of highly polymorphic inhibitory receptors which are specific for classical HLA class-I molecules. Peripheral blood and freshly prepared tumor cell suspensions (n = 60) as well as control samples (n = 32) were investigated for the distribution, phenotype, and functional relevance of CD158ab/KIR2DL1,-2/3 expressing NK-cells in glioblastoma (GBM) patients. We found that GBM were scarcely infiltrated by NK-cells that preferentially expressed CD158ab/KIR2DL1,-2/3 as inhibitory receptors, displayed reduced levels of the activating receptors CD335/NKp46, CD226/DNAM-1, CD159c/NKG2C, and showed diminished capacity to produce IFN-γ and perforin. Functional hypoactivity of GBM-derived NK-cells persisted despite IL-2 preactivation. Blockade with a specific KIR2DL-1,2/3 monoclonal antibody reversed NK-cell inhibition and significantly enhanced degranulation and IFN-γ production of IL-2 preactivated NK-cells in the presence of primary GBM cells and HLA-C expressing but not HLA class-I deficient K562 cells. Additional analysis revealed that significant amounts of IL-2 could be produced by tumor-derived CD4+ and CD8+CD45RA- memory T-cells after combined anti-CD3/anti-CD28 stimulation. Our data indicate that both blockade of inhibitory KIR and IL-2 triggering of tumor-derived NK-cells are necessary to enhance NK-cell responsiveness in GBM.
Collapse
Affiliation(s)
- Cüneyt Sönmez
- Department of Neurology With Institute of Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany.,Department of Spine Surgery, Klinikum Herford, 32049, Herford, Germany
| | - Johannes Wölfer
- Department of Neurosurgery, University Hospital Münster, Münster, Germany.,Department of Neurosurgery and Spine Surgery, Hufeland Klinikum GmbH, 99974, Mühlhausen, Germany
| | - Markus Holling
- Department of Neurosurgery, University Hospital Münster, Münster, Germany
| | - Benjamin Brokinkel
- Department of Neurosurgery, University Hospital Münster, Münster, Germany
| | - Walter Stummer
- Department of Neurosurgery, University Hospital Münster, Münster, Germany
| | - Heinz Wiendl
- Department of Neurology With Institute of Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany
| | - Christian Thomas
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Andreas Schulte-Mecklenbeck
- Department of Neurology With Institute of Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany
| | - Oliver M Grauer
- Department of Neurology With Institute of Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany.
| |
Collapse
|
37
|
Duan S, Liu S. Targeting NK Cells for HIV-1 Treatment and Reservoir Clearance. Front Immunol 2022; 13:842746. [PMID: 35371060 PMCID: PMC8967654 DOI: 10.3389/fimmu.2022.842746] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/07/2022] [Indexed: 12/31/2022] Open
Abstract
Combined antiretroviral therapy (cART) can inhibit the replication of human immunodeficiency virus type 1 (HIV-1) and reduce viral loads in the peripheral blood to undetectable levels. However, the presence of latent HIV-1 reservoirs prevents complete HIV-1 eradication. Several drugs and strategies targeting T cells are now in clinical trials, but their effectiveness in reducing viral reservoirs has been mixed. Interestingly, innate immune natural killer (NK) cells, which are promising targets for cancer therapy, also play an important role in HIV-1 infection. NK cells are a unique innate cell population with features of adaptive immunity that can regulate adaptive and innate immune cell populations; therefore, they can be exploited for HIV-1 immunotherapy and reservoir eradication. In this review, we highlight immunotherapy strategies for HIV infection that utilize the beneficial properties of NK cells.
Collapse
Affiliation(s)
- Siqin Duan
- Department of Clinical Laboratory, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Southern Medical University, Guangzhou, China
| |
Collapse
|
38
|
Rethacker L, Boy M, Bisio V, Roussin F, Denizeau J, Vincent-Salomon A, Borcoman E, Sedlik C, Piaggio E, Toubert A, Dulphy N, Caignard A. Innate lymphoid cells: NK and cytotoxic ILC3 subsets infiltrate metastatic breast cancer lymph nodes. Oncoimmunology 2022; 11:2057396. [PMID: 35371620 PMCID: PMC8973349 DOI: 10.1080/2162402x.2022.2057396] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Innate lymphoid cells (ILCs) – which include cytotoxic Natural Killer (NK) cells and helper-type ILC – are important regulators of tissue immune homeostasis, with possible roles in tumor surveillance. We analyzed ILC and their functionality in human lymph nodes (LN). In LN, NK cells and ILC3 were the prominent subpopulations. Among the ILC3s, we identified a CD56+/ILC3 subset with a phenotype close to ILC3 but also expressing cytotoxicity genes shared with NK. In tumor-draining LNs (TD-LNs) and tumor samples from breast cancer (BC) patients, NK cells were prominent, and proportions of ILC3 subsets were low. In tumors and TD-LN, NK cells display reduced levels of NCR (Natural cytotoxicity receptors), despite high transcript levels and included a small subset CD127− CD56− NK cells with reduced function. Activated by cytokines CD56+/ILC3 cells from donor and patients LN acquired cytotoxic capacity and produced IFNg. In TD-LN, all cytokine activated ILC populations produced TNFα in response to BC cell line. Analyses of cytotoxic and helper ILC indicate a switch toward NK cells in TD-LN. The local tumor microenvironment inhibited NK cell functions through downregulation of NCR, but cytokine stimulation restored their functionality.
Collapse
Affiliation(s)
- Louise Rethacker
- INSERM U1160, Institut de Recherche Saint-Louis, Hôpital Saint Louis, Paris, France
| | - Maxime Boy
- INSERM U1160, Institut de Recherche Saint-Louis, Hôpital Saint Louis, Paris, France
| | - Valeria Bisio
- INSERM U1160, Institut de Recherche Saint-Louis, Hôpital Saint Louis, Paris, France
| | - France Roussin
- Service d’Anesthésie-Réanimation, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Jordan Denizeau
- INSERM U932, Département de Recherche Translationelle, Institut Curie, Université de Recherche Paris Sciences & Lettres (PSL), Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Anne Vincent-Salomon
- Diagnostic and Theranostic Medicine Division, Institut Curie, PSL Research University, Paris, France
| | - Edith Borcoman
- Department of Medical Oncology, Institut Curie, Paris, France
- Université Paris Diderot, Université de Paris, Paris, France
| | - Christine Sedlik
- INSERM U932, Département de Recherche Translationelle, Institut Curie, Université de Recherche Paris Sciences & Lettres (PSL), Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Eliane Piaggio
- INSERM U932, Département de Recherche Translationelle, Institut Curie, Université de Recherche Paris Sciences & Lettres (PSL), Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Antoine Toubert
- INSERM U1160, Institut de Recherche Saint-Louis, Hôpital Saint Louis, Paris, France
- Université Paris Diderot, Université de Paris, Paris, France
- Assistance Publique–Hôpitaux de Paris (AP–HP), Hôpital Saint-Louis, Laboratoire d’Immunologie et Histocompatibilité, Paris, France
| | - Nicolas Dulphy
- INSERM U1160, Institut de Recherche Saint-Louis, Hôpital Saint Louis, Paris, France
- Université Paris Diderot, Université de Paris, Paris, France
- Assistance Publique–Hôpitaux de Paris (AP–HP), Hôpital Saint-Louis, Laboratoire d’Immunologie et Histocompatibilité, Paris, France
| | - Anne Caignard
- INSERM U1160, Institut de Recherche Saint-Louis, Hôpital Saint Louis, Paris, France
| |
Collapse
|
39
|
Allegra A, Petrarca C, Di Gioacchino M, Casciaro M, Musolino C, Gangemi S. Exosome-Mediated Therapeutic Strategies for Management of Solid and Hematological Malignancies. Cells 2022; 11:cells11071128. [PMID: 35406692 PMCID: PMC8997895 DOI: 10.3390/cells11071128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 12/24/2022] Open
Abstract
Exosomes are small membrane vesicles of endocytic origin containing cytokines, RNAs, growth factors, proteins, lipids, and metabolites. They have been identified as fundamental intercellular communication controllers in several diseases and an enormous volume of data confirmed that exosomes could either sustain or inhibit tumor onset and diffusion in diverse solid and hematological malignancies by paracrine signaling. Thus, exosomes might constitute a promising cell-free tumor treatment alternative. This review focuses on the effects of exosomes in the treatment of tumors, by discussing the most recent and promising data from in vitro and experimental in vivo studies and the few existing clinical trials. Exosomes are extremely promising as transporters of drugs, antagomir, genes, and other therapeutic substances that can be integrated into their core via different procedures. Moreover, exosomes can augment or inhibit non-coding RNAs, change the metabolism of cancer cells, and modify the function of immunologic effectors thus modifying the tumor microenvironment transforming it from pro-tumor to antitumor milieu. Here, we report the development of currently realized exosome modifiers that offer indications for the forthcoming elaboration of other more effective methods capable of enhancing the activity of the exosomes.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
- Correspondence: (A.A.); (M.D.G.)
| | - Claudia Petrarca
- Center for Advanced Studies and Technology, G. D’Annunzio University, 66100 Chieti, Italy;
- Department of Medicine and Aging Sciences, G. D’Annunzio University, 66100 Chieti, Italy
| | - Mario Di Gioacchino
- Center for Advanced Studies and Technology, G. D’Annunzio University, 66100 Chieti, Italy;
- Institute for Clinical Immunotherapy and Advanced Biological Treatments, 65100 Pescara, Italy
- Correspondence: (A.A.); (M.D.G.)
| | - Marco Casciaro
- Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, School of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy; (M.C.); (S.G.)
| | - Caterina Musolino
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Sebastiano Gangemi
- Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, School of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy; (M.C.); (S.G.)
| |
Collapse
|
40
|
Lettau M, Janssen O. Intra- and Extracellular Effector Vesicles From Human T And NK Cells: Same-Same, but Different? Front Immunol 2022; 12:804895. [PMID: 35003134 PMCID: PMC8733945 DOI: 10.3389/fimmu.2021.804895] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
Cytotoxic T lymphocytes (CTL) and Natural Killer (NK) cells utilize an overlapping effector arsenal for the elimination of target cells. It was initially proposed that all cytotoxic effector proteins are stored in lysosome-related effector vesicles (LREV) termed "secretory lysosomes" as a common storage compartment and are only released into the immunological synapse formed between the effector and target cell. The analysis of enriched LREV, however, revealed an uneven distribution of individual effectors in morphologically distinct vesicular entities. Two major populations of LREV were distinguished based on their protein content and signal requirements for degranulation. Light vesicles carrying FasL and 15 kDa granulysin are released in a PKC-dependent and Ca2+-independent manner, whereas dense granules containing perforin, granzymes and 9 kDa granulysin require Ca2+-signaling as a hallmark of classical degranulation. Notably, both types of LREV do not only contain the mentioned cytolytic effectors, but also store and transport diverse other immunomodulatory proteins including MHC class I and II, costimulatory and adhesion molecules, enzymes (i.e. CD26/DPP4) or cytokines. Interestingly, the recent analyses of CTL- or NK cell-derived extracellular vesicles (EV) revealed the presence of a related mixture of proteins in microvesicles or exosomes that in fact resemble fingerprints of the cells of origin. This overlapping protein profile indicates a direct relation of intra- and extracellular vesicles. Since EV potentially also interact with cells at distant sites (apart from the IS), they might act as additional effector vesicles or intercellular communicators in a more systemic fashion.
Collapse
Affiliation(s)
- Marcus Lettau
- Molecular Immunology, Institute of Immunology, University Hospital Schleswig-Holstein, Kiel, Germany.,Department of Internal Medicine II, Unit for Hematological Diagnostics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Ottmar Janssen
- Molecular Immunology, Institute of Immunology, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
41
|
Understanding natural killer cell biology from a single cell perspective. Cell Immunol 2022; 373:104497. [DOI: 10.1016/j.cellimm.2022.104497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/26/2022] [Accepted: 02/16/2022] [Indexed: 12/27/2022]
|
42
|
Kim N, Yi E, Kwon SJ, Park HJ, Kwon HJ, Kim HS. Filamin A Is Required for NK Cell Cytotoxicity at the Expense of Cytokine Production via Synaptic Filamentous Actin Modulation. Front Immunol 2022; 12:792334. [PMID: 35058930 PMCID: PMC8764188 DOI: 10.3389/fimmu.2021.792334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/13/2021] [Indexed: 11/23/2022] Open
Abstract
Natural killer (NK) cells are innate cytotoxic lymphocytes that efficiently eliminate malignant and virus-infected cells without prior activation via the directed and focused release of lytic granule contents for target cell lysis. This cytolytic process is tightly regulated at discrete checkpoint stages to ensure the selective killing of diseased target cells and is highly dependent on the coordinated regulation of cytoskeletal components. The actin-binding protein filamin crosslinks cortical actin filaments into orthogonal networks and links actin filament webs to cellular membranes to modulate cell migration, adhesion, and signaling. However, its role in the regulation of NK cell functions remains poorly understood. Here, we show that filamin A (FLNa), a filamin isoform with preferential expression in leukocytes, is recruited to the NK cell lytic synapse and is required for NK cell cytotoxicity through the modulation of conjugate formation with target cells, synaptic filamentous actin (F-actin) accumulation, and cytotoxic degranulation, but not granule polarization. Interestingly, we also find that the loss of FLNa augments the target cell-induced expression of IFN-γ and TNF-α by NK cells, correlating with enhanced activation signals such as Ca2+ mobilization, ERK, and NF-κB, and a delayed down-modulation of the NKG2D receptor. Thus, our results identify FLNa as a new regulator of NK cell effector functions during their decision to kill target cells through a balanced regulation of NK cell cytotoxicity vs cytokine production. Moreover, this study implicates the cross-linking/bundling of F-actin mediated by FLNa as a necessary process coordinating optimal NK effector functions.
Collapse
Affiliation(s)
- Nayoung Kim
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Eunbi Yi
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Soon Jae Kwon
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyo Jin Park
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyung-Joon Kwon
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hun Sik Kim
- Stem Cell Immunomodulation Research Center (SCIRC), University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
43
|
Zhang Z, Zhou Y, Lu J, Chen YF, Hu HY, Xu XQ, Fu GF. Changes in NK Cell Subsets and Receptor Expressions in HIV-1 Infected Chronic Patients and HIV Controllers. Front Immunol 2022; 12:792775. [PMID: 34975895 PMCID: PMC8716403 DOI: 10.3389/fimmu.2021.792775] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/30/2021] [Indexed: 01/31/2023] Open
Abstract
Natural killer (NK) cells are major effectors of the innate immune response and purported to play an influential role in the spontaneous control of HIV infection. In the present study, we compared the phenotypes of NK cells in the peripheral blood of three groups of subjects with chronic HIV-1 infection, HIV controllers, and healthy donors. The results showed that CD56+/CD16- NK cell subsets decreased in chronic patients and remained unchanged in controllers. Notably, we found that people living with chronic HIV-1 infection had suppressed NKp80, NKp46, and NKG2D expressions on NK cells compared to healthy donors, while HIV controllers remained unchanged. In contrast, NKG2D expression was substantially higher in controllers than in chronic patients (M=97.67, p<0.001). There were no significant differences in inhibitory receptors KIR3DL1 and KIR2DL1 expressions. In addition, plasma cytokine IFN-γ, TNF-α and IL-12showed higher levels in HIV controllers compared to chronic patients. Overall, our study revealed that, as compared to chronic patients, HIV controllers show an increased activating receptors expression and higher number ofCD56+/CD16-NK cell subset, with increased expression levels of plasma cytokines, suggesting that higher immune activation in controllers may have a key role in killing and suppressing HIV.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of HIV/STD Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Ying Zhou
- Department of HIV/STD Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Jing Lu
- Department of HIV/STD Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Yuan-Fang Chen
- Department of HIV/STD Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Hai-Yang Hu
- Department of HIV/STD Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Xiao-Qin Xu
- Department of HIV/STD Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Geng-Feng Fu
- Department of HIV/STD Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| |
Collapse
|
44
|
Gameiro SF, Evans AM, Mymryk JS. The tumor immune microenvironments of HPV + and HPV - head and neck cancers. WIREs Mech Dis 2022; 14:e1539. [PMID: 35030304 DOI: 10.1002/wsbm.1539] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/18/2021] [Accepted: 09/22/2021] [Indexed: 12/24/2022]
Abstract
Human papillomaviruses (HPVs) are the etiological agent of a significant, and increasing, fraction of head and neck squamous cell carcinomas (HNSCC)-a heterogenous group of malignancies in the head and neck region. HPV infection accounts for approximately 25% of all cases, with the remainder typically caused by smoking and excessive alcohol consumption. These distinct etiologies lead to profound clinical and immunological differences between HPV-positive (HPV+ ) and HPV-negative (HPV- ) HNSCC, likely related to the expression of exogenous viral antigens in the HPV+ subtype. Specifically, HPV+ HNSCC patients generally exhibit better treatment response compared to those with HPV- disease, leading to a more favorable prognosis, with lower recurrence rate, and longer overall survival time. Importantly, a plethora of studies have illustrated that the tumor immune microenvironment (TIME) of HPV+ HNSCC has a strikingly distinct immune composition to that of its HPV- counterpart. The HPV+ TIME is characterized as being immunologically "hot," with more immune infiltration, higher levels of T-cell activation, and higher levels of immunoregulation compared to the more immunologically "cold" HPV- TIME. In general, cancers with an immune "hot" TIME exhibit better treatment response and superior clinical outcomes in comparison to their immune "cold" counterparts. Indeed, this phenomenon has also been observed in HPV+ HNSCC patients, highlighting the critical role of the TIME in influencing prognosis, and further validating the use of cancer therapies that capitalize on the mobilization and/or modulation of the TIME. This article is categorized under: Cancer > Molecular and Cellular Physiology Infectious Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Steven F Gameiro
- Department of Microbiology and Immunology, The University of Western Ontario, London, Ontario, Canada
| | - Andris M Evans
- Department of Microbiology and Immunology, The University of Western Ontario, London, Ontario, Canada
| | - Joe S Mymryk
- Department of Microbiology and Immunology, The University of Western Ontario, London, Ontario, Canada.,Department of Otolaryngology, The University of Western Ontario, London, Ontario, Canada.,Department of Oncology, The University of Western Ontario, London, Ontario, Canada.,London Regional Cancer Program, Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
45
|
Cruz-Zárate D, Miguel-Rodríguez CE, Martínez-Vargas IU, Santos-Argumedo L. Myosin 1g and 1f: A Prospective Analysis in NK Cell Functions. Front Immunol 2022; 12:760290. [PMID: 34970258 PMCID: PMC8712487 DOI: 10.3389/fimmu.2021.760290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/23/2021] [Indexed: 02/05/2023] Open
Abstract
NK cells are contained in the ILC1 group; they are recognized for their antiviral and antitumor cytotoxic capacity; NK cells also participate in other immune response processes through cytokines secretion. However, the mechanisms that regulate these functions are poorly understood since NK cells are not as abundant as other lymphocytes, which has made them difficult to study. Using public databases, we identified that NK cells express mRNA encoding class I myosins, among which Myosin 1g and Myosin 1f are prominent. Therefore, this mini-review aims to generate a model of the probable participation of Myosin 1g and 1f in NK cells, based on information reported about the function of these myosins in other leukocytes.
Collapse
Affiliation(s)
- David Cruz-Zárate
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico.,Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Carlos Emilio Miguel-Rodríguez
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico.,Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Irving Ulises Martínez-Vargas
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico.,Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Leopoldo Santos-Argumedo
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| |
Collapse
|
46
|
Chakraborty S, Carnazza M, Jarboe T, DeSouza N, Li XM, Moscatello A, Geliebter J, Tiwari RK. Disruption of Cell-Cell Communication in Anaplastic Thyroid Cancer as an Immunotherapeutic Opportunity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1350:33-66. [PMID: 34888843 DOI: 10.1007/978-3-030-83282-7_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Thyroid cancer incidence is increasing at an alarming rate, almost tripling every decade. About 44,280 new cases of thyroid cancer (12,150 in men and 32,130 in women) are estimated to be diagnosed in 2021, with an estimated death toll of around 2200. Although most thyroid tumors are treatable and associated with a favorable outcome, anaplastic thyroid cancer (ATC) is extremely aggressive with a grim prognosis of 6-9 months post-diagnosis. A large contributing factor to this aggressive nature is that ATC is completely refractory to mainstream therapies. Analysis of the tumor microenvironment (TME) associated with ATC can relay insight to the pathological realm that encompasses tumors and aids in cancer progression and proliferation. The TME is defined as a complex niche that surrounds a tumor and involves a plethora of cellular components whose secretions can modulate the environment in order to favor tumor progression. The cellular heterogeneity of the TME contributes to its dynamic function due to the presence of both immune and nonimmune resident, infiltrating, and interacting cell types. Associated immune cells discussed in this chapter include macrophages, dendritic cells (DCs), natural killer (NK) cells, and tumor-infiltrating lymphocytes (TILs). Nonimmune cells also play a role in the establishment and proliferation of the TME, including neuroendocrine (NE) cells, adipocytes, endothelial cells (ECs), mesenchymal stem cells (MSCs), and fibroblasts. The dynamic nature of the TME contributes greatly to cancer progression.Recent work has found ATC tissues to be defined by a T cell-inflamed "hot" tumor immune microenvironment (TIME) as evidenced by presence of CD3+ and CD8+ T cells. These tumor types are amenable to immune checkpoint blockade (ICB) therapy. This therapeutic avenue, as of 2021, has remained unexplored in ATC. New studies should seek to explore the therapeutic feasibility of a combination therapy, through the use of a small molecule inhibitor with ICB in ATC. Screening of in vitro model systems representative of papillary, anaplastic, and follicular thyroid cancer explored the expression of 29 immune checkpoint molecules. There are higher expressions of HVEM, BTLA, and CD160 in ATC cell lines when compared to the other TC subtypes. The expression level of HVEM was more than 30-fold higher in ATC compared to the others, on average. HVEM is a member of tumor necrosis factor (TNF) receptor superfamily, which acts as a bidirectional switch through interaction with BTLA, CD160, and LIGHT, in a cis or trans manner. Given the T cell-inflamed hot TIME in ATC, expression of HVEM on tumor cells was suggestive of a possibility for complex crosstalk of HVEM with inflammatory cytokines. Altogether, there is emerging evidence of a T cell-inflamed TIME in ATC along with the expression of immune checkpoint proteins HVEM, BTLA, and CD160 in ATC. This can open doors for combination therapies using small molecule inhibitors targeting downstream effectors of MAPK pathway and antagonistic antibodies targeting the HVEM/BTLA axis as a potentially viable therapeutic avenue for ATC patients. With this being stated, the development of adaptive resistance to targeted therapies is inevitable; therefore, using a combination therapy that targets the TIME can serve as a preemptive tactic against the characteristic therapeutic resistance that is seen in ATC. The dynamic nature of the TME, including the immune cells, nonimmune cells, and acellular components, can serve as viable targets for combination therapy in ATC. Understanding the complex interactions of these associated cells and the paradigm in which their secretions and components can serve as immunomodulators are critical points of understanding when trying to develop therapeutics specifically tailored for the anaplastic thyroid carcinoma microenvironment.
Collapse
Affiliation(s)
- Sanjukta Chakraborty
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY, USA.,Weill Cornell Medicine, New York, NY, USA
| | - Michelle Carnazza
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY, USA
| | - Tara Jarboe
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY, USA
| | - Nicole DeSouza
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY, USA
| | - Xiu-Min Li
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY, USA
| | | | - Jan Geliebter
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY, USA
| | - Raj K Tiwari
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY, USA.
| |
Collapse
|
47
|
T Cell Subsets and Natural Killer Cells in the Pathogenesis of Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2021; 22:ijms222212190. [PMID: 34830072 PMCID: PMC8623596 DOI: 10.3390/ijms222212190] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/06/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a condition characterized by hepatic accumulation of excess lipids. T cells are commonly classified into various subsets based on their surface markers including T cell receptors, type of antigen presentation and pathophysiological functions. Several studies have implicated various T cell subsets and natural killer (NK) cells in the progression of NAFLD. While NK cells are mainly components of the innate hepatic immune system, the majority of T cell subsets can be part of both the adaptive and innate systems. Several studies have reported that various stages of NAFLD are accompanied by the accumulation of distinct T cell subsets and NK cells with different functions and phenotypes observed usually resulting in proinflammatory effects. More importantly, the overall stimulation of the intrahepatic T cell subsets is directly influenced by the homeostasis of the gut microbiota. Similarly, NK cells have been found to accumulate in the liver in response to pathogens and tumors. In this review, we discussed the nature and pathophysiological roles of T cell subsets including γδ T cells, NKT cells, Mucosal-associated invariant T (MAIT) cells as well as NK cells in NAFLD.
Collapse
|
48
|
Frazao A, Rethacker L, Jeudy G, Colombo M, Pasmant E, Avril MF, Toubert A, Moins-Teisserenc H, Roelens M, Dalac S, Maubec E, Caignard A. BRAF inhibitor resistance of melanoma cells triggers increased susceptibility to natural killer cell-mediated lysis. J Immunother Cancer 2021; 8:jitc-2019-000275. [PMID: 32912923 PMCID: PMC7482503 DOI: 10.1136/jitc-2019-000275] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2020] [Indexed: 12/11/2022] Open
Abstract
Background Targeted therapies and immunotherapies are first-line treatments for patients with advanced melanoma. Serine–threonine protein kinase B-RAF (BRAF) and mitogen-activated protein kinase (MEK) inhibition leads to a 70% response rate in patients with advanced melanoma with a BRAFV600E/K mutation. However, acquired resistance occurs in the majority of patients, leading to relapse. Immunotherapies that activate immune cytotoxic effectors induce long-lasting responses in 30% of patients. In that context, combination of targeted therapies with immunotherapy (IT) is a promising approach. We considered boosting natural killer (NK) cell tumor immunosurveillance, as melanoma cells express stress-induced molecules and activate NK cell lysis. Methods Here we have generated vemurafenib (a BRAF inihibitor)-resistant (R) cells from BRAFV600E SK28 and M14-sensitive (S) melanoma cell lines and investigated how resistance interferes with immunogenicity to NK cells. We determined the levels of several soluble molecules including NK ligands in 61 melanoma patients at baseline and 6 months M post-treatment with targeted therapies or immunotherapies. Results Vemurafenib resistance involved activation of p-AKT in SK28R and of p-MEK/p-ERK in M14R cells and was accompanied by modulation of NK ligands. Compared with S cells, SK28R displayed an increased expression of natural killer group 2 D (NKG2D) receptor ligands (major histocompatibility complex class (MHC) I chain-related protein A (MICA) and UL16-binding protein 2 (ULBP2)) whereas M14R exhibited decreased ULBP2. SK28R and M14R cells induced higher NK degranulation and interferon gamma secretion and were more efficiently lysed by donor and patient NK cells. SK28R showed increased tumor necrosis factor-related apoptosis-inducing ligand receptor II (TRAIL-RII) expression and TRAIL-induced apoptosis, and TRAIL-induced apoptosis of M14R was decreased. Combined BRAF/MEK inhibitors abrogated the growth of SK28S, M14S, and M14R cells, while growth of SK28R was maintained. BRAF/MEK inhibition attenuated NK activity but R cell lines activated polyfunctional NK cells and were lysed with high efficiency. We investigated the relationship of soluble NK ligands and response to treatment in a series of melanoma patients. Soluble NKG2D ligands known to regulate the receptor function have been associated to cancer progression. Serum analysis of patients treated with target therapies or IT indicates that soluble forms of NK ligands (MICA, B7H6, programmed cell death ligand 1, and carcinoembryonic antigen cell adhesion molecule 1) may correlate with clinical response. Conclusion These results support strategies combining targeted therapies and NK-based immunotherapies.
Collapse
Affiliation(s)
- Alexandra Frazao
- Université de Paris, INSERM UMRS-1160, Institut de Recherche Saint-Louis, 75010, Paris, France
| | - Louise Rethacker
- Université de Paris, INSERM UMRS-1160, Institut de Recherche Saint-Louis, 75010, Paris, France
| | - Géraldine Jeudy
- University Hospital Centre Dijon Bocage Complex, Dermatology Department, Dijon, France
| | - Marina Colombo
- Université de Paris, INSERM UMRS-1160, Institut de Recherche Saint-Louis, 75010, Paris, France
| | - Eric Pasmant
- Université de Paris, AP-HP Hôpital Cochin, Genetic and Molecular Biology Department, Institut Cochin, Paris, France
| | - Marie-Françoise Avril
- Université de Paris, AP-HP Hôpital Cochin, Dermatology Department, Institute Cochin, Paris, France
| | - Antoine Toubert
- Université de Paris, INSERM UMRS-1160, Institut de Recherche Saint-Louis, 75010, Paris, France
| | - Helene Moins-Teisserenc
- Université de Paris, INSERM UMRS-1160, AP-HP hopital Saint-Louis, Institut de Recherche Saint-Louis, 75010, Paris, France
| | - Marie Roelens
- Université de Paris, INSERM UMRS-1160, AP-HP hopital Saint-Louis, Institut de Recherche Saint-Louis, 75010, Paris, France
| | - Sophie Dalac
- University Hospital Centre Dijon Bocage Complex, Dermatology Department, Dijon, France
| | - Eve Maubec
- Université de Paris 13, AP-HP Hôpital Avicenne, Dermatology Department, Bobigny, France
| | - Anne Caignard
- Université de Paris, INSERM UMRS-1160, Institut de Recherche Saint-Louis, 75010, Paris, France
| |
Collapse
|
49
|
Jarahian M, Marstaller K, Banna N, Ahani R, Etemadzadeh MH, Boller LK, Azadmanesh K, Cid-Arregui A, Khezri A, Berger MR, Momburg F, Watzl C. Activating Natural Killer Cell Receptors, Selectins, and Inhibitory Siglecs Recognize Ebolavirus Glycoprotein. J Innate Immun 2021; 14:135-147. [PMID: 34425576 DOI: 10.1159/000517628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/28/2021] [Indexed: 11/19/2022] Open
Abstract
Expression of the extensively glycosylated Ebolavirus glycoprotein (EBOV-GP) induces physical alterations of surface molecules and plays a crucial role in viral pathogenicity. Here we investigate the interactions of EBOV-GP with host surface molecules using purified EBOV-GP, EBOV-GP-transfected cell lines, and EBOV-GP-pseudotyped lentiviral particles. Subsequently, we wanted to examine which receptors are involved in this recognition by binding studies to cells transfected with the EBOV-GP as well as to recombinant soluble EBOV-GP. As the viral components can also bind to inhibitory receptors of immune cells (e.g., Siglecs, TIM-1), they can even suppress the activity of immune effector cells. Our data show that natural killer (NK) cell receptors NKp44 and NKp46, selectins (CD62E/P/L), the host factors DC-SIGNR/DC-SIGN, and inhibitory Siglecs function as receptors for EBOV-GP. Our results show also moderate to strong avidity of homing receptors (P-, L-, and E-selectin) and DC-SIGNR/DC-SIGN to purified EBOV-GP, to cells transfected with EBOV-GP, as well as to the envelope of a pseudotyped lentiviral vector carrying the EBOV-GP. The concomitant activation and inhibition of the immune system exemplifies the evolutionary antagonism between the immune system and pathogens. Altogether these interactions with activating and inhibitory receptors result in a reduced NK cell-mediated lysis of EBOV-GP-expressing cells. Modulation of these interactions may provide new strategies for treating infections caused by this virus.
Collapse
Affiliation(s)
- Mostafa Jarahian
- Toxicology and Chemotherapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katharina Marstaller
- Toxicology and Chemotherapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nadine Banna
- Toxicology and Chemotherapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Roshanak Ahani
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Lea K Boller
- Department of Immunology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Dortmund, Germany
| | | | - Angel Cid-Arregui
- Targeted Tumor Vaccines Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Abdolrahman Khezri
- Department of Biotechnology, Inland Norway University of Applied Sciences, Hamar, Norway
| | - Martin R Berger
- Toxicology and Chemotherapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Momburg
- Antigen Presentation and T/NK Cell Activation Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carsten Watzl
- Department of Immunology, Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Dortmund, Germany
| |
Collapse
|
50
|
Fortes-Andrade T, Almeida JS, Sousa LM, Santos-Rosa M, Freitas-Tavares P, Casanova JM, Rodrigues-Santos P. The Role of Natural Killer Cells in Soft Tissue Sarcoma: Prospects for Immunotherapy. Cancers (Basel) 2021; 13:cancers13153865. [PMID: 34359767 PMCID: PMC8345358 DOI: 10.3390/cancers13153865] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Soft-tissue sarcomas (STS) represent about 80% of sarcomas, and are a heterogeneous group of rare and malignant tumors. Morphological evaluation has been the standard model for the diagnosis of sarcomas, and even in samples with similar characteristics, they present genetic differences, which further increases the diversity of sarcomas. This variety is one of the main challenges for the classification and understanding of STS patterns, as well as for the respective treatments, which further decreases patient survival (<5 years). Natural Killer (NK) cells have a fundamental role in the control and immune surveillance of cancer development, progression and metastases. Notwithstanding the scarcity of studies to characterize NK cells in STS, it is noteworthy that the progression of these malignancies is associated with altered NK cells. These findings support the additional need to explore NK cell-based immunotherapy in STS; some clinical trials, although very tentatively, are already underway. Abstract Soft-tissue sarcomas (STS) represent about 80% of sarcomas, and are a heterogeneous group of rare and malignant tumors. STS arise from mesenchymal tissues and can grow into structures such as adipose tissue, muscles, nervous tissue and blood vessels. Morphological evaluation has been the standard model for the diagnosis of sarcomas, and even in samples with similar characteristics, they present a diversity in cytogenetic and genetic sequence alterations, which further increases the diversity of sarcomas. This variety is one of the main challenges for the classification and understanding of STS patterns, as well as for their respective treatments, which further decreases patient survival (<5 years). Despite some studies, little is known about the immunological profile of STS. As for the immunological profile of STS in relation to NK cells, there is also a shortage of studies. Observations made in solid tumors show that the infiltration of NK cells in tumors is associated with a good prognosis of the disease. Notwithstanding the scarcity of studies to characterize NK cells, their receptors, and ligands in STS, it is noteworthy that the progression of these malignancies is associated with altered NK phenotypes. Despite the scarcity of information on the function of NK cells, their phenotypes and their regulatory pathways in STS, the findings of this study support the additional need to explore NK cell-based immunotherapy in STS further. Some clinical trials, very tentatively, are already underway. STS clinical trials are still the basis for adoptive NK-cell and cytokine-based therapy.
Collapse
Affiliation(s)
- Tânia Fortes-Andrade
- Center for Neuroscience and Cell Biology (CNC), Laboratory of Immunology and Oncology, University of Coimbra, 3004-504 Coimbra, Portugal; (T.F.-A.); (J.S.A.); (L.M.S.)
| | - Jani Sofia Almeida
- Center for Neuroscience and Cell Biology (CNC), Laboratory of Immunology and Oncology, University of Coimbra, 3004-504 Coimbra, Portugal; (T.F.-A.); (J.S.A.); (L.M.S.)
- Faculty of Medicine, Immunology Institute, University of Coimbra, 3004-504 Coimbra, Portugal;
- Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Luana Madalena Sousa
- Center for Neuroscience and Cell Biology (CNC), Laboratory of Immunology and Oncology, University of Coimbra, 3004-504 Coimbra, Portugal; (T.F.-A.); (J.S.A.); (L.M.S.)
| | - Manuel Santos-Rosa
- Faculty of Medicine, Immunology Institute, University of Coimbra, 3004-504 Coimbra, Portugal;
- Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Paulo Freitas-Tavares
- Coimbra Hospital and University Center (CHUC), Tumor Unit of the Locomotor Apparatus (UTAL), University Clinic of Orthopedics, Orthopedics Service, 3000-075 Coimbra, Portugal;
| | - José Manuel Casanova
- Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Hospital and University Center (CHUC), Tumor Unit of the Locomotor Apparatus (UTAL), University Clinic of Orthopedics, Orthopedics Service, 3000-075 Coimbra, Portugal;
| | - Paulo Rodrigues-Santos
- Center for Neuroscience and Cell Biology (CNC), Laboratory of Immunology and Oncology, University of Coimbra, 3004-504 Coimbra, Portugal; (T.F.-A.); (J.S.A.); (L.M.S.)
- Faculty of Medicine, Immunology Institute, University of Coimbra, 3004-504 Coimbra, Portugal;
- Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Correspondence: ; Tel.: +351-239-85-77-77 (ext. 24-28-44)
| |
Collapse
|